1
|
Yu Y, Wang Y, Zhang J, Bu Q, Jiang D, Jiang Y, Xu L, Ju Z, Chen T. Anaerobic probiotics-in situ Se nanoradiosensitizers selectively anchor to tumor with immuno-regulations for robust cancer radio-immunotherapy. Biomaterials 2025; 318:123117. [PMID: 39864125 DOI: 10.1016/j.biomaterials.2025.123117] [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: 08/07/2024] [Revised: 01/06/2025] [Accepted: 01/18/2025] [Indexed: 01/28/2025]
Abstract
Developing translational nanoradiosensitizers with multiple activities in sensitizing tumor cells and re-shaping tumor immunosuppressive microenvironments are urgently desired for addressing the poor therapeutic efficacy of radiotherapy in clinic. Inspired by the anaerobic and immunoagonist properties of the probiotic (bifidobacterium longum, BL), herein, a biomimetic Selenium nanoradiosensitizer in situ-formed on the surface of the probiotic (BL@SeNPs) is developed in a facile method to potentiate radiotherapy. BL@SeNPs selectively target to hypoxia regions of tumors and then anchor on the surface of tumor cells to inhibit its proliferation. Meanwhile, it also significantly promotes ROS generations to damage DNA and induces cell cycle arrest for enhancing the therapeutic efficacy of radiotherapy, which will induce immunogenic cell death to initiate antitumor immunities. In addition, BL@SeNPs nanoradiosensitizers can serve as immunoagonist to activate immune cells like dendritic cells (DCs) to further magnify the quality of the induced immune responses. More importantly, BL@SeNPs combining radiotherapy effectively reduce immunosuppressor cells (e.g. TAM, MDSC, TAN) infiltrating within tumors for shaping tumor microenvironments to effectively combat tumor progressions. This study provides a safe, effective and translational nanoradiosensitizer and its combination radiotherapy for clinical cancer treatment and shed lights for developing next generation of nanoradiosensitizers.
Collapse
Affiliation(s)
- Yangyang Yu
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China
| | - Ying Wang
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China
| | - Jin Zhang
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China
| | - Qingyue Bu
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China
| | - Dan Jiang
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China
| | - Yalin Jiang
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China
| | - Ligeng Xu
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangdong, 510632, China.
| | - Tianfeng Chen
- Department of Pharmacy of Puning People's Hospital (Guangdong Postdoctoral Innovation Practice Base of Jinan University), Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangdong, 510632, China.
| |
Collapse
|
2
|
Luo Q, Bai X, Li X, Liu C. The role and mechanism of selenium in the prevention and progression of hepatocellular carcinoma. Front Oncol 2025; 15:1557233. [PMID: 40182029 PMCID: PMC11965637 DOI: 10.3389/fonc.2025.1557233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025] Open
Abstract
Hepatocellular carcinoma (HCC) represents the most prevalent form of liver cancer. Despite notable advancements in therapeutic strategies, HCC continues to pose significant public health challenges due to its rising incidence and high mortality rates worldwide. Selenium is an essential trace element that playing a critical role in human health. Recent studies have highlighted its potential preventive and therapeutic benefits in the context of HCC. However, some in vitro and in vivo investigations have yielded inconsistent results, and the mechanisms by which selenium influences HCC are still not completely clear. This review begins by providing an extensive evaluation of the effects and mechanisms of selenium on the primary risk factors associated with HCC, including viral infections, metabolic abnormalities, and lifestyle factors. Subsequently, we outline the roles and mechanisms by which selenium influences the proliferation, metastasis, and immune microenvironment of HCC. Finally, we emphasize the imperative for further investigation into the optimal dosage and forms of selenium, as well as its effects on the HCC microenvironment, to inform the development of effective clinical strategies. This review thus provides a foundational framework for the potential clinical application of selenium in the treatment of HCC.
Collapse
Affiliation(s)
- Qinying Luo
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaofang Bai
- Department of Ultrasonography, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Xiaojiao Li
- BioBank, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Chang Liu
- Department of Clinical Laboratory, Gongli Hospital of Shanghai Pudong, Shanghai, China
| |
Collapse
|
3
|
Cai L, Chen H, Wang Y, Zhang J, Song D, Tan Y, Guo Z, Wang X. Platinum(IV) Complexes Trigger Death Receptors and Natural Killer Cells to Suppress Breast Cancer. J Med Chem 2025. [PMID: 39886904 DOI: 10.1021/acs.jmedchem.4c02509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Chemoimmunotherapy is an alternative treatment against cancers. Platinum(IV) complexes FMP and DFMP, coupling formononetin derivative as axial ligand(s), were designed to suppress triple-negative breast cancer (TNBC) by activating death receptors (DRs) and natural killer (NK) cells. These complexes show great potential to overcome the resistance of TNBC to chemotherapy by inducing both intrinsic and extrinsic apoptosis in cancer cells. Particularly, FMP with one axial formononetin derivative not only induced the caspase-3-dependent intrinsic apoptosis but also upregulated the expression of DRs and caspase-8, triggered the extrinsic apoptosis, and enhanced the cytotoxic ability of NK92 cells. Moreover, FMP increased the release of granzyme B, restrained the proliferation and differentiation of myeloid-derived suppressor cells, and the secretion of IL-10, thus inhibiting the TNBC in vitro and in vivo. The results demonstrate that FMP overcomes the chemoresistance and immune escape of TNBC through a new mechanism involving the synergy of chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Linxiang Cai
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Hanhua Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Ying Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jingwen Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Dongfan Song
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Yehong Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
4
|
Ali BA, Allam RM, Hasanin MS, Hassabo AA. Biosynthesis of selenium nanoparticles as a potential therapeutic agent in breast cancer: G2/M arrest and apoptosis induction. Toxicol Rep 2024; 13:101792. [PMID: 39554610 PMCID: PMC11565031 DOI: 10.1016/j.toxrep.2024.101792] [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: 08/27/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/19/2024] Open
Abstract
The drawbacks and adverse reactions of conventional breast cancer (BC) medications have prompted researchers to seek novel therapeutic approaches. This study aimed to study the impact of biosynthesized selenium nanoparticles by yeast on breast cancer (MCF-7) cells and to find potential underlying mechanisms. Therefore, marine yeast isolates were screened for their ability to biosynthesis selenium nanoparticles (SeNPs). The most potent isolate was identified as Candida pseudojiufengensis based on 18 S rRNA gene sequencing. Incubation of cell-free extract with 0.8 mM of SeO2 for 48 h at 40°C in pH of 7.0 were optimal conditions for the biosynthesis of SeNPs. The biosynthesized SeNPs were characterized by UV-Vis spectroscopy, X-ray diffraction (XRD), transmission electron microscopy (TEM), and dynamic light scattering (DLS) measurements including average particle size distribution and average zeta potential. The results showed that the biosynthesized SeNPs displayed a maximum absorbance peak in the UV-Vis spectrum at 560 nm due to surface plasmon resonance. TEM image elevated spherical shape particles with an average size of 12 nm. SRB assay, flow cytometry, and other biochemical methods were employed to assess SeNPs anti-proliferative effects on MCF-7 cells. SeNPs showed superior anticancer efficacy against MCF-7 cells compared to colon (HCT-116) and liver (HepG2) cancer cells, as evidenced by lower IC50 values (19.59 µg/ml) against 36.36 µg/ml and 27.81 ±1.4 µg/ml, respectively. However, SeNPs demonstrated no cytotoxic effects against HSF cells. Moreover, treatment with SeNPs induces G2/M arrest along with triggering apoptosis in MCF-7 cells. Furthermore, MCF-7 cells treated with SeNPs showed increased oxidative stress, as indicated by observable rises in LPO and 8-OHDG, accompanied by considerable exhaustion in antioxidant enzyme activity. These findings demonstrated that Se nanoparticles synthesized from yeast have therapeutic promise in BC treatment.
Collapse
Affiliation(s)
- Basant A. Ali
- Microbial Chemistry Department, Biotechology Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Rasha Mosa Allam
- Pharmacology Department, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Mohamed S. Hasanin
- Cellulose and Paper Department, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Amany A. Hassabo
- Microbial Chemistry Department, Biotechology Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| |
Collapse
|
5
|
Maleczek M, Reszeć-Giełażyn J, Szymulewska-Konopko K. Beneficial Effects of Selenium and Its Supplementation on Carcinogenesis and the Use of Nanoselenium in the Treatment of Malignant Tumors. Int J Mol Sci 2024; 25:11285. [PMID: 39457066 PMCID: PMC11508626 DOI: 10.3390/ijms252011285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Selenium was recognized as a non-toxic element in the second half of the 20th century. Since then, the positive impact of selenium on the functioning of the human body has been noticed. It has been shown that low levels of selenium in the body are significantly associated with a higher risk of developing cancer. Selenium acts as an antioxidant and inhibits the proliferation of cancer cells. It has been shown that selenium supplementation may contribute to reducing the risk of DNA mutations and carcinogenesis. Nanomedicine has become very helpful in both the diagnosis and treatment of cancer. Due to its anticancer properties, selenium is used in nanotechnology as selenium nanoparticles.
Collapse
Affiliation(s)
| | - Joanna Reszeć-Giełażyn
- Department of Medical Pathomorphology, Medical University of Białystok, 15-269 Białystok, Poland; (M.M.)
| | | |
Collapse
|
6
|
Chen L, Xu YX, Wang YS, Ren YY, Dong XM, Wu P, Xie T, Zhang Q, Zhou JL. Prostate cancer microenvironment: multidimensional regulation of immune cells, vascular system, stromal cells, and microbiota. Mol Cancer 2024; 23:229. [PMID: 39395984 PMCID: PMC11470719 DOI: 10.1186/s12943-024-02137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is one of the most prevalent malignancies in males worldwide. Increasing research attention has focused on the PCa microenvironment, which plays a crucial role in tumor progression and therapy resistance. This review aims to provide a comprehensive overview of the key components of the PCa microenvironment, including immune cells, vascular systems, stromal cells, and microbiota, and explore their implications for diagnosis and treatment. METHODS Keywords such as "prostate cancer", "tumor microenvironment", "immune cells", "vascular system", "stromal cells", and "microbiota" were used for literature retrieval through online databases including PubMed and Web of Science. Studies related to the PCa microenvironment were selected, with a particular focus on those discussing the roles of immune cells, vascular systems, stromal cells, and microbiota in the development, progression, and treatment of PCa. The selection criteria prioritized peer-reviewed articles published in the last five years, aiming to summarize and analyze the latest research advancements and clinical relevance regarding the PCa microenvironment. RESULTS The PCa microenvironment is highly complex and dynamic, with immune cells contributing to immunosuppressive conditions, stromal cells promoting tumor growth, and microbiota potentially affecting androgen metabolism. Vascular systems support angiogenesis, which fosters tumor expansion. Understanding these components offers insight into the mechanisms driving PCa progression and opens avenues for novel therapeutic strategies targeting the tumor microenvironment. CONCLUSIONS A deeper understanding of the PCa microenvironment is crucial for advancing diagnostic techniques and developing precision therapies. This review highlights the potential of targeting the microenvironment to improve patient outcomes, emphasizing its significance in the broader context of PCa research and treatment innovation.
Collapse
Affiliation(s)
- Lin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yu-Xin Xu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Yuan-Shuo Wang
- School of Pharmacy, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Ying-Ying Ren
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xue-Man Dong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Pu Wu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Qi Zhang
- Department of Urology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310014, China.
| | - Jian-Liang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
7
|
Bu Q, Jiang D, Yu Y, Deng Y, Chen T, Xu L. Surface chemistry engineered selenium nanoparticles as bactericidal and immuno-modulating dual-functional agents for combating methicillin-resistant Staphylococcus aureus Infection. Drug Resist Updat 2024; 76:101102. [PMID: 38936006 DOI: 10.1016/j.drup.2024.101102] [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: 01/28/2024] [Revised: 05/26/2024] [Accepted: 06/09/2024] [Indexed: 06/29/2024]
Abstract
Because of the extremely complexed microenvironment of drug-resistant bacterial infection, nanomaterials with both bactericidal and immuno-modulating activities are undoubtedly the ideal modality for overcoming drug resistance. Herein, we precisely engineered the surface chemistry of selenium nanoparticles (SeNPs) using neutral (polyvinylpyrrolidone-PVP), anionic (letinan-LET) and cationic (chitosan-CS) surfactants. It was found that surface chemistry greatly influenced the bioactivities of functionalized SeNPs, their interactions with methicillin-resistant Staphylococcus aureus (MRSA), immune cells and metabolisms. LET-functionalized SeNPs with distinct metabolisms exhibited the best inhibitory efficacy compared to other kinds of SeNPs against MRSA through inducing robust ROS generation and damaging bacterial cell wall. Meanwhile, only LET-SeNPs could effectively activate natural kill (NK) cells, and enhance the phagocytic capability of macrophages and its killing activity against bacteria. Furthermore, in vivo studies suggested that LET-SeNPs treatment highly effectively combated MRSA infection and promoted wound healing by triggering much more mouse NK cells, CD8+ and CD4+ T lymphocytes infiltrating into the infected area at the early stage to efficiently eliminate MRSA in the mouse model. This study demonstrates that the novel functionalized SeNP with dual functions could serve as an effective antibacterial agent and could guide the development of next generation antibacterial agents.
Collapse
Affiliation(s)
- Qingyue Bu
- Department of Intensive Care Unit, The First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou 510632, China
| | - Dan Jiang
- Department of Intensive Care Unit, The First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou 510632, China
| | - Yangyang Yu
- Department of Intensive Care Unit, The First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou 510632, China
| | - Yunqing Deng
- Department of Intensive Care Unit, The First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Intensive Care Unit, The First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou 510632, China.
| | - Ligeng Xu
- Department of Intensive Care Unit, The First Affiliated Hospital, Department of Chemistry, State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Viral Pathogenesis & Infection Prevention and Control, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
8
|
Kostić M, Marjanović J, Divac V. Organoselenium transition metal complexes as promising candidates in medicine area. J Biol Inorg Chem 2024; 29:555-571. [PMID: 39123093 DOI: 10.1007/s00775-024-02072-y] [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: 05/09/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
The medicinal properties of transition metal complexes are greatly influenced by the nature and physico-chemical features of the ligand present in the complex structure. Due to the unique biological properties of the organoselenium compounds reflected in the variety of pharmacological activities (such as antioxidative, antiviral, antimicrobial and anticancer), the last years have brought increased interest for their use as a ligands compounds in the design and syntheses of range of transition metal-based coordination compounds that have been explored as antitumor and antimicrobial agents. Our aim in this review is to provide the overview of an recent development of the transition metal complexes bearing organoselenium ligands in the structure that could be promising choice for the treatment of various diseases, particularly cancer and infective diseases. For this purpose, the complexes of Co, Ni, Cu, Zn, Ru, Pd, Pt, Au and Sn as the most explored examples will be included and discussed.
Collapse
Affiliation(s)
- Marina Kostić
- Institute for Information Technologies, University of Kragujevac, Jovana Cvijića bb, 34000, Kragujevac, Serbia.
| | - Jovana Marjanović
- Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Vera Divac
- Faculty of Science, University of Kragujevac, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| |
Collapse
|
9
|
Gao Y, Huang D, Huang S, Li H, Xia B. Rational design of ROS generation nanosystems to regulate innate immunity of macrophages, dendrtical and natural killing cells for immunotherapy. Int Immunopharmacol 2024; 139:112695. [PMID: 39024751 DOI: 10.1016/j.intimp.2024.112695] [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: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Innate immunity serves as the first line of host defense in the body against pathogenic infections or malignant diseases. Reactive oxygen species (ROS), as vital signaling mediators, can efficiently elicit innate immune responses to oxidative-related stress or damage. In the era of nanomedicine, various immunostimulatory nanosystems have been extensively designed and synthesized to elicit immune responses for the immunotherapy of cancer or infectious diseases. In this review, we emphasize that ROS derived from nanosystems regulates innate immune cells to potentiate immunotherapeutic efficacy, such as primarily dendritic cells, macrophages, or natural killer cells. Meanwhile, we also summarize the pathway of ROS generation triggered by exogenous nanosystems in innate immune cells of DCs, macrophages, and NK cells.
Collapse
Affiliation(s)
- Yan Gao
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China
| | - Di Huang
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China
| | - Shuodan Huang
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China
| | - Huiying Li
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, PR China.
| | - Bing Xia
- College of Science, State Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, PR China; Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, PR China.
| |
Collapse
|
10
|
Liu C, Wang W, Lai H, Chen Y, Li L, Li H, Zhan M, Chen T, Cao W, Li X. Biosynthesis of fungus-based oral selenium microcarriers for radioprotection and immuno-homeostasis shaping against radiation-induced heart disease. Bioact Mater 2024; 37:393-406. [PMID: 38689659 PMCID: PMC11059443 DOI: 10.1016/j.bioactmat.2024.03.034] [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: 12/23/2023] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Abstract
Radiation-induced heart disease (RIHD), characterized by severe oxidative stress and immune dysregulation, is a serious condition affecting cancer patients undergoing thoracic radiation. Unfortunately, clinical interventions for RIHD are lacking. Selenium (Se) is a trace element with excellent antioxidant and immune-modulatory properties. However, its application in heart radioprotection remains challenging. Herein, we developed a novel bioactive Cordyceps militaris-based Se oral delivery system (Se@CM), which demonstrated superior radioprotection effects in vitro against X-ray-induced damage in H9C2 cells through suppressing excessive ROS generation, compared to the radioprotectant Amifostine. Moreover, Se@CM exhibited exceptional cardioprotective effects in vivo against X-ray irradiation, reducing cardiac dysfunction and myocardial fibrosis by balancing the redox equilibrium and modulating the expression of Mn-SOD and MDA. Additionally, Se@CM maintained immuno-homeostasis, as evidenced by the upregulated population of T cells and M2 macrophages through modulation of selenoprotein expression after irradiation. Together, these results highlight the remarkable antioxidant and immunity modulation properties of Se@CM and shed light on its promising application for cardiac protection against IR-induced disease. This research provides valuable insights into developing effective strategies for preventing and managing RIHD.
Collapse
Affiliation(s)
- Chang Liu
- Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, PR China
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd, Jinan University, Zhuhai 519000, China
| | - Weiyi Wang
- Department of Chemistry, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Haoqiang Lai
- Department of Chemistry, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Yikang Chen
- Guangdong Jinan Established Selenium Source Nano Technology Research Institute Co., Ltd., Guangzhou 510535, China
| | - Lvyi Li
- Department of Chemistry, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Haiwei Li
- Guangdong Jinan Established Selenium Source Nano Technology Research Institute Co., Ltd., Guangzhou 510535, China
| | - Meixiao Zhan
- Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, PR China
| | - Tianfeng Chen
- Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, 519000, PR China
- Department of Chemistry, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Wenqiang Cao
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd, Jinan University, Zhuhai 519000, China
| | - Xiaoling Li
- Department of Chemistry, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| |
Collapse
|
11
|
Hu R, Lan J, Zhang D, Shen W. Nanotherapeutics for prostate cancer treatment: A comprehensive review. Biomaterials 2024; 305:122469. [PMID: 38244344 DOI: 10.1016/j.biomaterials.2024.122469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Prostate cancer (PCa) is the most prevalent solid organ malignancy and seriously affects male health. The adverse effects of prostate cancer therapeutics can cause secondary damage to patients. Nanotherapeutics, which have special targeting abilities and controlled therapeutic release profiles, may serve as alternative agents for PCa treatment. At present, many nanotherapeutics have been developed to treat PCa and have shown better treatment effects in animals than traditional therapeutics. Although PCa nanotherapeutics are highly attractive, few successful cases have been reported in clinical practice. To help researchers design valuable nanotherapeutics for PCa treatment and avoid useless efforts, herein, we first reviewed the strategies and challenges involved in prostate cancer treatment. Subsequently, we presented a comprehensive review of nanotherapeutics for PCa treatment, including their targeting methods, controlled release strategies, therapeutic approaches and mechanisms. Finally, we proposed the future prospects of nanotherapeutics for PCa treatment.
Collapse
Affiliation(s)
- Ruimin Hu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jin Lan
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Dinglin Zhang
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Wenhao Shen
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
12
|
Xu Y, Lai H, Pan S, Pan L, Liu T, Yang Z, Chen T, Zhu X. Selenium promotes immunogenic radiotherapy against cervical cancer metastasis through evoking P53 activation. Biomaterials 2024; 305:122452. [PMID: 38154440 DOI: 10.1016/j.biomaterials.2023.122452] [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: 10/14/2023] [Revised: 12/06/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
Radiotherapy is still the recommended treatment for cervical cancer. However, radioresistance and radiation-induced side effects remain one of the biggest clinical problems. Selenium (Se) has been confirmed to exhibit radiation-enhancing effects for cancer treatment. However, Se species dominate the biological activities and which form of Se possesses better radiosensitizing properties and radiation safety remains elusive. Here, different Se species (the valence state of Se ranged from - 2, 0, +4 to + 6) synergy screen was carried out to identify the potential radiosensitizing effects and radiation safety of Se against cervical cancer. We found that the therapeutic effects varied with the changes in the Se valence state. Sodium selenite (+4) displayed strong cancer-killing effects but also possessed severe cytotoxicity. Sodium selenate (+6) neither enhanced the killing effects of X-ray nor possessed anticancer activity by its alone treatment. Although nano-selenium (0), especially Let-SeNPs, has better radiosensitizing activity, the - 2 organic Se, such as selenadiazole derivative SeD (-2) exhibited more potent anticancer effects and possessed a higher safe index. Overall, the selected Se drugs were able to synergize with X-ray to inhibit cell growth, clone formation, and cell migration by triggering G2/M phase arrest and apoptosis, and SeD (-2) was found to exhibit more potent enhancing capacity. Further mechanism studies showed that SeD mediated p53 pathway activation by inducing DNA damage through promoting ROS production. Additionally, SeD combined with X-ray therapy can induce an anti-tumor immune response in vivo. More importantly, SeD combined with X-ray significantly inhibited the liver metastasis of tumor cells and alleviated the side effects caused by radiation therapy in tumor-bearing mice. Taken together, this study demonstrates the radiosensitization and radiation safety effects of different Se species, which may shed light on the application of such Se-containing drugs serving as side effects-reducing agents for cervical cancer radiation treatment.
Collapse
Affiliation(s)
- Yanchao Xu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China; Department of Chemistry, Jinan University, China
| | - Haoqiang Lai
- Department of Chemistry, Jinan University, China
| | - Shuya Pan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Liuliu Pan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Ting Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Ziyi Yang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China
| | - Tianfeng Chen
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China; Department of Chemistry, Jinan University, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, China.
| |
Collapse
|
13
|
Zhou C, Lu Z, Sun B, Yi Y, Zhang B, Wang Z, Qiu SJ. Peripheral Lymphocytes in Primary Liver Cancers: Elevated NK and CD8+ T Cells and Dysregulated Selenium Metabolism. Biomolecules 2024; 14:222. [PMID: 38397459 PMCID: PMC10886987 DOI: 10.3390/biom14020222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Peripheral blood lymphocytes (PBLs), which play a pivotal role in orchestrating the immune system, garner minimal attention in hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). The impact of primary liver cancers on PBLs remains unexplored. In this study, flow cytometry facilitated the quantification of cell populations, while transcriptome of PBLs was executed utilizing 10× single-cell sequencing technology. Additionally, pertinent cases were curated from the GEO database. Subsequent bioinformatics and statistical analyses were conducted utilizing R (4.2.1) software. Elevated counts of NK cells and CD8+ T cells were observed in both ICC and HCC when compared to benign liver disease (BLD). In the multivariate Cox model, NK cells and CD8+ T cells emerged as independent risk factors for recurrence-free survival. Single-cell sequencing of PBLs uncovered the downregulation of TGFβ signaling in tumor-derived CD8+ T cells. Pathway enrichment analysis, based on differential expression profiling, highlighted aberrations in selenium metabolism. Proteomic analysis of preoperative and postoperative peripheral blood samples from patients undergoing tumor resection revealed a significant upregulation of SELENBP1 and a significant downregulation of SEPP1. Primary liver cancer has a definite impact on PBLs, manifested by alterations in cellular quantities and selenoprotein metabolism.
Collapse
Affiliation(s)
- Cheng Zhou
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (C.Z.); (B.S.)
| | - Zhufeng Lu
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, China;
| | - Baoye Sun
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (C.Z.); (B.S.)
| | - Yong Yi
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (C.Z.); (B.S.)
| | - Boheng Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (C.Z.); (B.S.)
- Department of Hepatic Oncology, Xiamen Clinical Research Center for Cancer Therapy, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen 361015, China
- Center for Evidence-Based Medicine, Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Zheng Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (C.Z.); (B.S.)
| | - Shuang-Jian Qiu
- Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (C.Z.); (B.S.)
| |
Collapse
|
14
|
Himoto T, Masaki T. Current Trends on the Involvement of Zinc, Copper, and Selenium in the Process of Hepatocarcinogenesis. Nutrients 2024; 16:472. [PMID: 38398797 PMCID: PMC10892613 DOI: 10.3390/nu16040472] [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: 12/11/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Numerous nutritional factors increase the risk of hepatocellular carcinoma (HCC) development. The dysregulation of zinc, copper, and selenium homeostasis is associated with the occurrence of HCC. The impairment of the homeostasis of these essential trace elements results in oxidative stress, DNA damage, cell cycle progression, and angiogenesis, finally leading to hepatocarcinogenesis. These essential trace elements can affect the microenvironment in HCC. The carrier proteins for zinc and copper and selenium-containing enzymes play important roles in the prevention or progression of HCC. These trace elements enhance or alleviate the chemosensitivity of anticancer agents in patients with HCC. The zinc, copper, or selenium may affect the homeostasis of other trace elements with each other. Novel types of cell death including ferropotosis and cupropotosis are also associated with hepatocarcinogenesis. Therapeutic strategies for HCC that target these carrier proteins for zinc and copper or selenium-containing enzymes have been developed in in vitro and in vivo studies. The use of zinc-, copper- or selenium-nanoparticles has been considered as novel therapeutic agents for HCC. These results indicate that zinc, copper, and selenium may become promising therapeutic targets in patients with HCC. The clinical application of these agents is an urgent unmet requirement. This review article highlights the correlation between the dysregulation of the homeostasis of these essential trace elements and the development of HCC and summarizes the current trends on the roles of these essential trace elements in the pathogenesis of hepatocarcinogenesis.
Collapse
Affiliation(s)
- Takashi Himoto
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1, Hara, Mure-cho, Takamatsu 761-0123, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1, Ikenobe, Miki-cho 761-0793, Kagawa, Japan
| |
Collapse
|
15
|
Park J, Kim S, Jangid AK, Park HW, Kim K. Networked Cluster Formation via Trigonal Lipid Modules for Augmented Ex Vivo NK Cell Priming. Int J Mol Sci 2024; 25:1556. [PMID: 38338836 PMCID: PMC10855780 DOI: 10.3390/ijms25031556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Current cytokine-based natural killer (NK) cell priming techniques have exhibited limitations such as the deactivation of biological signaling molecules and subsequent insufficient maturation of the cell population during mass cultivation processes. In this study, we developed an amphiphilic trigonal 1,2-distearoyl-sn-glycero-3-phosphorylethanolamine (DSPE) lipid-polyethylene glycol (PEG) material to assemble NK cell clusters via multiple hydrophobic lipid insertions into cellular membranes. Our lipid conjugate-mediated ex vivo NK cell priming sufficiently augmented the structural modulation of clusters, facilitated diffusional signal exchanges, and finally activated NK cell population with the clusters. Without any inhibition in diffusional signal exchanges and intrinsic proliferative efficacy of NK cells, effectively prime NK cell clusters produced increased interferon-gamma, especially in the early culture periods. In conclusion, the present study demonstrates that our novel lipid conjugates could serve as a promising alternative for future NK cell mass production.
Collapse
Affiliation(s)
| | | | | | | | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul 22012, Republic of Korea; (J.P.); (S.K.); (A.K.J.); (H.W.P.)
| |
Collapse
|
16
|
Jangid AK, Kim S, Park HW, Kim HJ, Kim K. Ex Vivo Surface Decoration of Phenylboronic Acid onto Natural Killer Cells for Sialic Acid-Mediated Versatile Cancer Cell Targeting. Biomacromolecules 2024; 25:222-237. [PMID: 38130077 DOI: 10.1021/acs.biomac.3c00916] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Phenylboronic acid (PBA) has been highly acknowledged as a significant cancer recognition moiety in sialic acid-overexpressing cancer cells. In this investigation, lipid-mediated biomaterial integrated PBA molecules onto the surface of natural killer (NK) cells to make a receptor-mediated immune cell therapeutic module. Therefore, a 1,2-distearoyl-sn-glycero-3-phosphorylethanolamine (DSPE) lipid-conjugated di-PEG-PBA (DSPEPEG-di(PEG-PBA) biomaterial was synthesized. The DSPEPEG-di(PEG-PBA) biomaterial exhibited a high affinity for sialic acid (SA), confirmed by fluorescence spectroscopy at pH 6.5 and 7.4. DSPEPEG-di(PEG-PBA) was successfully anchored onto NK cell surfaces (PBA-NK), and this biomaterial maintains intrinsic properties such as viability, ligand availability (FasL & TRAIL), and cytokine secretion response to LPS. The anticancer efficacy of PBA-NK cells was evaluated against 2D cancer cells (MDA-MB-231, HepG2, and HCT-116) and 3D tumor spheroids of MDA-MB-231 cells. PBA-NK cells exhibited greatly enhanced anticancer effects against SA-overexpressing cancer cells. Thus, PBA-NK cells represent a new anticancer strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Hee Won Park
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| | - Hyun Jin Kim
- Department of Biological Engineering, College of Engineering, Inha University, Incheon 22212, South Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul 04620, South Korea
| |
Collapse
|
17
|
Zhang H, Yang L, Wang T, Li Z. NK cell-based tumor immunotherapy. Bioact Mater 2024; 31:63-86. [PMID: 37601277 PMCID: PMC10432724 DOI: 10.1016/j.bioactmat.2023.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/16/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
Natural killer (NK) cells display a unique inherent ability to identify and eliminate virus-infected cells and tumor cells. They are particularly powerful for elimination of hematological cancers, and have attracted considerable interests for therapy of solid tumors. However, the treatment of solid tumors with NK cells are less effective, which can be attributed to the very complicated immunosuppressive microenvironment that may lead to the inactivation, insufficient expansion, short life, and the poor tumor infiltration of NK cells. Fortunately, the development of advanced nanotechnology has provided potential solutions to these issues, and could improve the immunotherapy efficacy of NK cells. In this review, we summarize the activation and inhibition mechanisms of NK cells in solid tumors, and the recent advances in NK cell-based tumor immunotherapy boosted by diverse nanomaterials. We also propose the challenges and opportunities for the clinical application of NK cell-based tumor immunotherapy.
Collapse
Affiliation(s)
- Hao Zhang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Li Yang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Tingting Wang
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| | - Zhen Li
- Center for Molecular Imaging and Nuclear Medicine, State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, 215123, China
| |
Collapse
|
18
|
Lee CE, Kim S, Park HW, Lee W, Jangid AK, Choi Y, Jeong WJ, Kim K. Tailoring tumor-recognizable hyaluronic acid-lipid conjugates to enhance anticancer efficacies of surface-engineered natural killer cells. NANO CONVERGENCE 2023; 10:56. [PMID: 38097911 PMCID: PMC10721593 DOI: 10.1186/s40580-023-00406-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023]
Abstract
Natural killer (NK) cells have clinical advantages in adoptive cell therapy owing to their inherent anticancer efficacy and their ability to identify and eliminate malignant tumors. However, insufficient cancer-targeting ligands on NK cell surfaces often inhibit their immunotherapeutic performance, especially in immunosuppressive tumor microenvironment. To facilitate tumor recognition and subsequent anticancer function of NK cells, we developed hyaluronic acid (HA, ligands to target CD44 overexpressed onto cancer cells)-poly(ethylene glycol) (PEG, cytoplasmic penetration blocker)-Lipid (molecular anchor for NK cell membrane decoration through hydrophobic interaction) conjugates for biomaterial-mediated ex vivo NK cell surface engineering. Among these major compartments (i.e., Lipid, PEG and HA), optimization of lipid anchors (in terms of chemical structure and intrinsic amphiphilicity) is the most important design parameter to modulate hydrophobic interaction with dynamic NK cell membranes. Here, three different lipid types including 1,2-dimyristoyl-sn-glycero-3-phosphati-dylethanolamine (C14:0), 1,2-distearoyl-sn-glycero-3-phosphatidylethanolamine (DSPE, C18:0), and cholesterol were evaluated to maximize membrane coating efficacy and associated anticancer performance of surface-engineered NK cells (HALipid-NK cells). Our results demonstrated that NK cells coated with HA-PEG-DSPE conjugates exhibited significantly enhanced anticancer efficacies toward MDA-MB-231 breast cancer cells without an off-target effect on human fibroblasts specifically via increased NK cell membrane coating efficacy and prolonged surface duration of HA onto NK cell surfaces, thereby improving HA-CD44 recognition. These results suggest that our HALipid-NK cells with tumor-recognizable HA-PEG-DSPE conjugates could be further utilized in various cancer immunotherapies.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Hee Won Park
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Wonjeong Lee
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea
| | - Yonghyun Choi
- Department of Chemical Science and Engineering, Tokyo Institute of Technology, Kanagawa, 226-8501, Japan
| | - Woo-Jin Jeong
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea.
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, 04620, Republic of Korea.
| |
Collapse
|
19
|
Zhu M, Liu D, Liu G, Zhang M, Pan F. Caspase-Linked Programmed Cell Death in Prostate Cancer: From Apoptosis, Necroptosis, and Pyroptosis to PANoptosis. Biomolecules 2023; 13:1715. [PMID: 38136586 PMCID: PMC10741419 DOI: 10.3390/biom13121715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/08/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Prostate cancer (PCa) is a complex disease and the cause of one of the highest cancer-related mortalities in men worldwide. Annually, more than 1.2 million new cases are diagnosed globally, accounting for 7% of newly diagnosed cancers in men. Programmed cell death (PCD) plays an essential role in removing infected, functionally dispensable, or potentially neoplastic cells. Apoptosis is the canonical form of PCD with no inflammatory responses elicited, and the close relationship between apoptosis and PCa has been well studied. Necroptosis and pyroptosis are two lytic forms of PCD that result in the release of intracellular contents, which induce inflammatory responses. An increasing number of studies have confirmed that necroptosis and pyroptosis are also closely related to the occurrence and progression of PCa. Recently, a novel form of PCD named PANoptosis, which is a combination of apoptosis, necroptosis, and pyroptosis, revealed the attached connection among them and may be a promising target for PCa. Apoptosis, necroptosis, pyroptosis, and PANoptosis are good examples to better understand the mechanism underlying PCD in PCa. This review aims to summarize the emerging roles and therapeutic potential of apoptosis, necroptosis, pyroptosis, and PANoptosis in PCa.
Collapse
Affiliation(s)
- Minggang Zhu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.Z.); (D.L.); (M.Z.)
| | - Di Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.Z.); (D.L.); (M.Z.)
| | - Guoqiang Liu
- Urology Department of Guangzhou First People’s Hospital, Guangzhou 510000, China;
| | - Mingrui Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.Z.); (D.L.); (M.Z.)
| | - Feng Pan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.Z.); (D.L.); (M.Z.)
| |
Collapse
|
20
|
Deng B, He X, Wang D, Wang Y, Jiang Y, Chen T, Xu L. Designing Selenium Nanoadjuvant as Universal Agent for Live-Killed Virus-Based Vaccine. SMALL METHODS 2023; 7:e2300293. [PMID: 37491791 DOI: 10.1002/smtd.202300293] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/01/2023] [Indexed: 07/27/2023]
Abstract
Inactivated virus vaccines with whole antigen spectra and good safety are the commonly used modality for preventing infections. However, the poor immunogenicity greatly limits its clinical applications. Herein, by taking advantages of the crucial roles of Se in the functions of immune cells and its biomineralization property, it successfully in-situ synthesized Se nanoadjuvant on inactivated viruses such as porcine epidemic diarrhea virus (PEDV), pseudorabies virus (PRV), and porcine reproductive and respiratory syndrome virus (PRRSV) in a facile method, which is universal to construct other inactivated virus vaccines. The nanovaccine can highly effectively enhance the uptake of PEDV/PRV/PRRSV into dendritic cells (DCs) and activate DCs via triggering TLR4 signaling pathways and regulating selenoproteins expressions. Furthermore, it exhibited better activities in triggering macrophages and natural killer cells-mediated innate immunity and T cells-mediated cellular immunity compared to PEDV and the commercial inactivated PEDV vaccine on both mice and swine models. This study provides a universal Se nanoadjuvant for developing inactivated viruses-based nanovaccines for preventing virus infections.
Collapse
Affiliation(s)
- Bo Deng
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaoming He
- Wens Foodstuff Group Co. Ltd, Yunfu, Guangdong, 527400, China
| | - Dongdong Wang
- Wens Foodstuff Group Co. Ltd, Yunfu, Guangdong, 527400, China
| | - Ying Wang
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Yalin Jiang
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Tianfeng Chen
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Ligeng Xu
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, Guangdong, 510632, China
| |
Collapse
|
21
|
Lavrova MA, Verzun SA, Mishurinskiy SA, Sirotin MA, Bykova SK, Gontcharenko VE, Mariasina SS, Korshunov VM, Taydakov IV, Belousov YA, Dolzhenko VD. Fine-Tuning of the Optical and Electrochemical Properties of Ruthenium(II) Complexes with 2-Arylbenzimidazoles and 4,4'-Dimethoxycarbonyl-2,2'-bipyridine. Molecules 2023; 28:6541. [PMID: 37764316 PMCID: PMC10536653 DOI: 10.3390/molecules28186541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
A series of cyclometalated complexes of ruthenium (II) with four different substituents in the aryl fragment of benzimidazole was synthesized in order to study the effect of substituent donation on the electronic structure of the substances. The resulting complexes were studied using X-ray diffraction, NMR spectroscopy, MALDI mass spectrometry, electron absorption spectroscopy, luminescence spectroscopy, and cyclic voltammetry as well as DFT/TDDFT was also used to interpret the results. All the complexes have intense absorption in the range of up to 700 nm, the triplet nature of the excited state was confirmed by measurement of luminescence decay. With an increase in substituent donation, a red shift of the absorption and emission bands occurs, and the lifetime of the excited state and the redox potential of the complex decrease. The combination of these properties shows that the complexes are excellent dyes and can be used as photosensitizers.
Collapse
Affiliation(s)
- Maria A. Lavrova
- Chemistry Department, M.V. Lomonosov Moscow State University, Leninskie Gory Street, Building 1/3, 119234 Moscow, Russia; (S.A.V.); (S.A.M.); (M.A.S.); (S.S.M.); (Y.A.B.)
| | - Stepan A. Verzun
- Chemistry Department, M.V. Lomonosov Moscow State University, Leninskie Gory Street, Building 1/3, 119234 Moscow, Russia; (S.A.V.); (S.A.M.); (M.A.S.); (S.S.M.); (Y.A.B.)
| | - Sergey A. Mishurinskiy
- Chemistry Department, M.V. Lomonosov Moscow State University, Leninskie Gory Street, Building 1/3, 119234 Moscow, Russia; (S.A.V.); (S.A.M.); (M.A.S.); (S.S.M.); (Y.A.B.)
| | - Maxim A. Sirotin
- Chemistry Department, M.V. Lomonosov Moscow State University, Leninskie Gory Street, Building 1/3, 119234 Moscow, Russia; (S.A.V.); (S.A.M.); (M.A.S.); (S.S.M.); (Y.A.B.)
- N.N. Semenov Federal Research Center for Chemical Physics, Kosygina Street 4, 119991 Moscow, Russia
| | - Sofya K. Bykova
- Higher Chemical College of RAS, Mendeleev University of Chemical Technology, Miusskaya Square, 9, 125047 Moscow, Russia;
| | - Victoria E. Gontcharenko
- Faculty of Chemistry, National Research University Higher School of Economics, 20 Miasnitskaya Street, 101000 Moscow, Russia;
- P.N. Lebedev Physical Institute of the Russian Academy of Sciences, 119991 Moscow, Russia; (V.M.K.); (I.V.T.)
| | - Sofia S. Mariasina
- Chemistry Department, M.V. Lomonosov Moscow State University, Leninskie Gory Street, Building 1/3, 119234 Moscow, Russia; (S.A.V.); (S.A.M.); (M.A.S.); (S.S.M.); (Y.A.B.)
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia
- Institute of Functional Genomics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Vladislav M. Korshunov
- P.N. Lebedev Physical Institute of the Russian Academy of Sciences, 119991 Moscow, Russia; (V.M.K.); (I.V.T.)
- Faculty of Fundamental Sciences, Bauman Moscow State Technical University, 105005 Moscow, Russia
| | - Ilya V. Taydakov
- P.N. Lebedev Physical Institute of the Russian Academy of Sciences, 119991 Moscow, Russia; (V.M.K.); (I.V.T.)
- Academic Department of Innovational Materials and Technologies Chemistry, G.V. Plekhanov Russian University of Economics, 36 Stremyannoy per., 117997 Moscow, Russia
| | - Yury A. Belousov
- Chemistry Department, M.V. Lomonosov Moscow State University, Leninskie Gory Street, Building 1/3, 119234 Moscow, Russia; (S.A.V.); (S.A.M.); (M.A.S.); (S.S.M.); (Y.A.B.)
- P.N. Lebedev Physical Institute of the Russian Academy of Sciences, 119991 Moscow, Russia; (V.M.K.); (I.V.T.)
| | - Vladimir D. Dolzhenko
- Chemistry Department, M.V. Lomonosov Moscow State University, Leninskie Gory Street, Building 1/3, 119234 Moscow, Russia; (S.A.V.); (S.A.M.); (M.A.S.); (S.S.M.); (Y.A.B.)
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky pr. 47, 119991 Moscow, Russia
| |
Collapse
|
22
|
Jiang J, Chen B, Tang B, Wei Q. Selenium in Prostate Cancer: Prevention, Progression, and Treatment. Pharmaceuticals (Basel) 2023; 16:1250. [PMID: 37765058 PMCID: PMC10536940 DOI: 10.3390/ph16091250] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Selenium, a trace mineral with various biological functions, has become a focal point in prostate cancer research. This review aims to present a comprehensive overview of selenium's involvement in prostate cancer, covering its impact on prevention, development, treatment, and underlying mechanisms. Observational studies have revealed a link between selenium levels and selenoproteins with prostate cancer progression. However, randomized controlled studies have shown that selenium supplementation does not prevent prostate cancer (HR: 0.95; 95% CI 0.80-1.13). This discrepancy might be attributed to selenoprotein single nucleotide polymorphisms. In the context of combinatorial therapy, selenium has demonstrated promising synergistic potential in the treatment of prostate cancer. Emerging evidence highlights the significant role of selenium and selenoproteins in prostate cancer, encompassing AR signaling, antioxidative properties, cell death, cell cycle regulation, angiogenesis, epigenetic regulation, immunoregulation, epithelial-mesenchymal transformation, and redox signal. In conclusion, selenium's diverse properties make it a promising trace mineral in prostate cancer prevention, development, and treatment and as a platform for exploring novel agents.
Collapse
Affiliation(s)
- Jinjiang Jiang
- Department of Urology, West China Hospital of Sichuan University, No. 37, Guoxue Lane, Chengdu 610041, China
- Institute of Urology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Bo Chen
- Department of Urology, West China Hospital of Sichuan University, No. 37, Guoxue Lane, Chengdu 610041, China
- Institute of Urology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Bo Tang
- Department of Urology, West China Hospital of Sichuan University, No. 37, Guoxue Lane, Chengdu 610041, China
- Institute of Urology, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Qiang Wei
- Department of Urology, West China Hospital of Sichuan University, No. 37, Guoxue Lane, Chengdu 610041, China
- Institute of Urology, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
23
|
Shao J, Jin Y, Jin C. A new approach to overcoming resistance to immunotherapy: nanotechnology. Front Oncol 2023; 13:1210245. [PMID: 37637050 PMCID: PMC10457008 DOI: 10.3389/fonc.2023.1210245] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Immunotherapy for immune response has ushered in a new era of cancer treatment. In recent years, new immunotherapeutic agents have been introduced into clinical trials and even approved for marketing. However, the widespread use of immunotherapeutic agents faces an unavoidable challenge: immunotherapy does not work at all for some patients, or has good efficacy in the initial phase, but immunotherapy resistance develops within a short period of time, and immunotherapy can also cause serious adverse effects such as autoimmune inflammation and non-specific inflammation. How to enable patients to overcome drug resistance, reduce the toxic side effects of drugs, enhance patient compliance and improve patient survival has become a problem that clinicians have to face. The advent of nanotechnology provides an encouraging platform for immunotherapy. It can not only improve the bioavailability and stability of drugs and reduce toxic side effects, but also reduce resistance to immunotherapy. Here, we discuss these research advances and discuss potential challenges and future directions.
Collapse
Affiliation(s)
- Jiangbo Shao
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Jin
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chunxiang Jin
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
24
|
Khawar MB, Gao G, Rafiq M, Shehzadi A, Afzal A, Abbasi MH, Sheikh N, Afzal N, Ashraf MA, Hamid SE, Shahzaman S, Kawish N, Sun H. Breaking down barriers: The potential of smarter CAR-engineered NK cells against solid tumors. J Cell Biochem 2023; 124:1082-1104. [PMID: 37566723 DOI: 10.1002/jcb.30460] [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: 03/30/2023] [Revised: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Natural killer (NK) cells are considered to be the foremost fighters of our innate immune system against foreign invaders and thus tend to promptly latch onto the virus-infected and tumor/cancerous cells, killing them through phagocytosis. At present, the application of genetically engineered Chimeric antigen receptor (CAR) receptors ensures a guaranteed optimistic response with NK cells and would not allow the affected cells to dodge or escape unchecked. Hence the specificity and uniqueness of CAR-NK cells over CAR-T therapy make them a better immunotherapeutic choice to reduce the load of trafficking of numerous tumor cells near the healthy cell populations in a more intact way than offered by CAR-T immunotherapy. Our review mainly focuses on the preclinical, clinical, and recent advances in clinical research trials and further strategies to achieve an augmented and efficient cure against solid tumors.
Collapse
Affiliation(s)
- Muhammad B Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
- Department of Zoology, Applied Molecular Biology and Biomedicine Lab, University of Narowal, Narowal, Pakistan
| | - Guangzhong Gao
- Department of Physiatry, Haian Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nantong, Jiangsu, China
| | - Mussarat Rafiq
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Anila Shehzadi
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Ali Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Nadeem Sheikh
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Nimra Afzal
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | | | - Syeda E Hamid
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Sara Shahzaman
- Department of Zoology, Molecular Medicine and Cancer Therapeutics Lab, University of Central Punjab, Lahore, Pakistan
| | - Naseer Kawish
- Cell & Molecular Biology Lab, Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental and, Translational Noncoding RNA Research, Yangzhou, China
| |
Collapse
|
25
|
Alfadul SM, Matnurov EM, Varakutin AE, Babak MV. Metal-Based Anticancer Complexes and p53: How Much Do We Know? Cancers (Basel) 2023; 15:2834. [PMID: 37345171 DOI: 10.3390/cancers15102834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/23/2023] Open
Abstract
P53 plays a key role in protecting the human genome from DNA-related mutations; however, it is one of the most frequently mutated genes in cancer. The P53 family members p63 and p73 were also shown to play important roles in cancer development and progression. Currently, there are various organic molecules from different structural classes of compounds that could reactivate the function of wild-type p53, degrade or inhibit mutant p53, etc. It was shown that: (1) the function of the wild-type p53 protein was dependent on the presence of Zn atoms, and (2) Zn supplementation restored the altered conformation of the mutant p53 protein. This prompted us to question whether the dependence of p53 on Zn and other metals might be used as a cancer vulnerability. This review article focuses on the role of different metals in the structure and function of p53, as well as discusses the effects of metal complexes based on Zn, Cu, Fe, Ru, Au, Ag, Pd, Pt, Ir, V, Mo, Bi and Sn on the p53 protein and p53-associated signaling.
Collapse
Affiliation(s)
- Samah Mutasim Alfadul
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Egor M Matnurov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Alexander E Varakutin
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Hong Kong SAR 999077, China
| |
Collapse
|
26
|
Liu S, Wei W, Wang J, Chen T. Theranostic applications of selenium nanomedicines against lung cancer. J Nanobiotechnology 2023; 21:96. [PMID: 36935493 PMCID: PMC10026460 DOI: 10.1186/s12951-023-01825-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/18/2023] [Indexed: 03/21/2023] Open
Abstract
The incidence and mortality rates of lung cancer are among the highest in the world. Traditional treatment methods include surgery, chemotherapy, and radiotherapy. Although rapid progress has been achieved in the past decade, treatment limitations remain. It is therefore imperative to identify safer and more effective therapeutic methods, and research is currently being conducted to identify more efficient and less harmful drugs. In recent years, the discovery of antitumor drugs based on the essential trace element selenium (Se) has provided good prospects for lung cancer treatments. In particular, compared to inorganic Se (Inorg-Se) and organic Se (Org-Se), Se nanomedicine (Se nanoparticles; SeNPs) shows much higher bioavailability and antioxidant activity and lower toxicity. SeNPs can also be used as a drug delivery carrier to better regulate protein and DNA biosynthesis and protein kinase C activity, thus playing a role in inhibiting cancer cell proliferation. SeNPs can also effectively activate antigen-presenting cells to stimulate cell immunity, exert regulatory effects on innate and regulatory immunity, and enhance lung cancer immunotherapy. This review summarizes the application of Se-based species and materials in lung cancer diagnosis, including fluorescence, MR, CT, photoacoustic imaging and other diagnostic methods, as well as treatments, including direct killing, radiosensitization, chemotherapeutic sensitization, photothermodynamics, and enhanced immunotherapy. In addition, the application prospects and challenges of Se-based drugs in lung cancer are examined, as well as their forecasted future clinical applications and sustainable development.
Collapse
Affiliation(s)
- Shaowei Liu
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Weifeng Wei
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jinlin Wang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Tianfeng Chen
- College of Chemistry and Materials Science, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
27
|
Zhao R, Miao W, Li B. The Relation Between Trace Elements and Latent Tuberculosis Infection: a Study Based on National Health and Nutritional Examination Survey 2011-2012. Biol Trace Elem Res 2023; 201:1080-1089. [PMID: 35482174 DOI: 10.1007/s12011-022-03240-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/05/2022] [Indexed: 02/07/2023]
Abstract
This study aimed to analyze the potential association between trace elements and latent tuberculosis infection (LTBI) based on the data from the National Health and Nutritional Examination Survey (NHANES) during 2011-2012. In this cross-sectional study, tuberculin skin testing (TST) and QuantiFERON®-TB Gold In-Tube (QFT-GIT) were utilized to screen for LTBI. Participants with positive results of TST or/and QFT-GIT were defined as LTBI. Weighted univariate and multivariate logistic regression analyses were used to explore the association between trace elements and LTBI. Subgroup analyses were conducted according to gender, age, birthplace, race, and health insurance holding status. A total of 6064 participants were included in this study, of whom 655 (10.80%) participants were with positive results of LTBI. Weighted multivariable analysis demonstrated that zinc [odds ratio (OR) = 0.89; 95% confidence interval (CI), 0.82-0.97] and selenium (OR = 0.31; 95%CI, 0.13-0.70) in the serum may be associated with a reduced risk of LTBI. In different concentrations of zinc and selenium, serum zinc concentration of 12.56-13.99 μmol/l (vs. < 11.23 μmol/l; OR = 0.37, 95% CI, 0.20-0.67) was related to a reduced risk of LTBI, while no significant difference was observed under different selenium levels (P > 0.05). Subgroup analyses indicated that the role of zinc and selenium in reducing TB risk may be more significant in males, people aged 21-64, people born in the USA, people with health insurance, and non-Hispanic Whites. Maintaining serum zinc and selenium levels may help reduce the risk of LTBI and indirectly help people prevent TB.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Respiratory and Critical Care Medicine, Chifeng Municipal Hospital, Chifeng, 024000, People's Republic of China
| | - Wei Miao
- Department of Endocrinology, Chifeng Municipal Hospital, Chifeng, 024000, People's Republic of China
| | - Baohua Li
- Department of Infectious Disease, Dingxi People's Hospital, No. 22 Anding Road, Dingxi, 743000, People's Republic of China.
| |
Collapse
|
28
|
Yang Y, Jiang Y, Xie B, Shi S, Pi F, Chen M, Sang C, Xu L, Chen T. Selenadiazole derivative-loaded metal azolate frameworks facilitate NK cell immunotherapy by sensitizing tumor cells and shaping immuno-suppressive microenvironments. Biomater Sci 2023; 11:1517-1529. [PMID: 36606484 DOI: 10.1039/d2bm01752k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The low sensitivity of tumor cells and immunosuppressive microenvironments lead to unsatisfactory efficacy of natural killer (NK) cell immunotherapy. In this work, we developed a safe and effective combination treatment strategy by integrating a selenadiazole derivative (PSeD)-loaded metal azolate framework (PSeD@MAF-4(R)) with NK cells derived from cancer patients against a xenograft human breast tumor model. Intriguingly, it was found that only PSeD@MAF-4(R) pretreatment on tumor cells exhibited synergistic effects with NK cells in inhibiting tumor cell growth by up-regulating NKG2D and its ligands to maximize the interactions between NK and MCF-7 cells. Moreover, PSeD@MAF-4(R) pretreatment could significantly enhance the degranulation of NK cells and regulate their secretions of pro- or anti-inflammatory cytokines (e.g. IL-6, IL-10, and TGF-β). Furthermore, PSeD@MAF-4(R) could significantly enhance the penetration capability of NK cells into tumor spheroids. The combination treatment mainly induced G1 phase arrest and activated multiple caspase-mediated apoptosis of tumor cells. In vivo evidence showed that PSeD@MAF-4(R) combined with NK cells could highly efficiently combat breast tumor progression via inducing and activating innate immune cell (DC and NK cell) infiltrations within tumor tissues while shaping the suppressive tumor microenvironment by down-regulating the expression of TGF-β. This developed strategy may provide important information for developing NK cell-based combination cancer immunotherapy with high efficacy and good safety profiles.
Collapse
Affiliation(s)
- Yahui Yang
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Yalin Jiang
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Bin Xie
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Sujiang Shi
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Fen Pi
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Mingkai Chen
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Chengcheng Sang
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Ligeng Xu
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| | - Tianfeng Chen
- Department of Chemistry, Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
29
|
Nie S, He X, Sun Z, Zhang Y, Liu T, Chen T, Zhao J. Selenium speciation-dependent cancer radiosensitization by induction of G2/M cell cycle arrest and apoptosis. Front Bioeng Biotechnol 2023; 11:1168827. [PMID: 37034255 PMCID: PMC10073679 DOI: 10.3389/fbioe.2023.1168827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction: Radiation therapy has Q6long been a routine and effective treatment for non-small cell lung cancer (NSCLC), but the radioresistance and side effects have limited its application. In recent years, the superiority showed by trace element selenium in tumor radiotherapy sensitization has received wide attention. However, different forms of selenium compounds exhibit different chemical properties and their mechanisms of action on tumors may be different. Methods: Human non-small cell lung cancer SPC-A1 cells were studied. Drug toxicity was detected by MTT assay. The selenium content absorbed in vitro at different time points was detected by ICP-MS. Colony formation were conducted to observe the radiosensitization effect of different selenium compounds on SPC-A1 cells, and to compare the proliferation ability of SPC-A1 cells treated by radiation alone and radiation combined with different selenium compounds. Cell migration was detected by cell scratch assay. The changes of cell cycle and apoptosis were detected by flow cytometry. DCFH-DA fluorescent probe was used to detect the effects of different selenium compounds combined with X-ray on ROS production. Results: In this study, these four representative selenium compounds all have a certain ability to enhance the ability of radiotherapy to inhibit tumor cell proliferation and migration, and the mechanism may be related to blocking cell cycle in G2/M phase, activating the caspase cascade and reducing intracellular ROS levels to induce tumor cell apoptosis. Among them, -2-valent organic selenium has the most obvious effect, mainly inhibits cell migration, and induces early apoptosis by activating a large number of caspase-3, and arrest the cell cycle in S phase and G2/M phase. 0-valent selenium nanoparticles mainly arrest the cell cycle in G2/M phase. +4-valent inorganic selenium exerts its antitumor effects primarily by inhibiting tumor cell migration and inducing early apoptosis of tumor cells. Discussion: In this paper, the antitumor effects of four different forms of selenium compounds combined with X-rays on SPC-A1 cells were investigated, and their inhibitory effects on the proliferation and migration of cancer cells and their mechanisms were examined. We found that the radiosensitizing effect of selenium on NSCLC was closely related to its selenium form through the study of the sensitizing effect of different kinds of selenium compounds on radiotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jianfu Zhao
- *Correspondence: Tianfeng Chen, ; Jianfu Zhao,
| |
Collapse
|
30
|
Garbo S, Di Giacomo S, Łażewska D, Honkisz-Orzechowska E, Di Sotto A, Fioravanti R, Zwergel C, Battistelli C. Selenium-Containing Agents Acting on Cancer-A New Hope? Pharmaceutics 2022; 15:pharmaceutics15010104. [PMID: 36678733 PMCID: PMC9860877 DOI: 10.3390/pharmaceutics15010104] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/18/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
Selenium-containing agents are more and more considered as an innovative potential treatment option for cancer. Light is shed not only on the considerable advancements made in understanding the complex biology and chemistry related to selenium-containing small molecules but also on Se-nanoparticles. Numerous Se-containing agents have been widely investigated in recent years in cancer therapy in relation to tumour development and dissemination, drug delivery, multidrug resistance (MDR) and immune system-related (anti)cancer effects. Despite numerous efforts, Se-agents apart from selenocysteine and selenomethionine have not yet reached clinical trials for cancer therapy. The purpose of this review is to provide a concise critical overview of the current state of the art in the development of highly potent target-specific Se-containing agents.
Collapse
Affiliation(s)
- Sabrina Garbo
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Silvia Di Giacomo
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Dorota Łażewska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688 Kraków, Poland
| | - Ewelina Honkisz-Orzechowska
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College in Kraków, Medyczna 9, 30-688 Kraków, Poland
| | - Antonella Di Sotto
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Rossella Fioravanti
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
- Correspondence: (C.Z.); (C.B.)
| | - Cecilia Battistelli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Correspondence: (C.Z.); (C.B.)
| |
Collapse
|
31
|
Ramos-Inza S, Plano D, Sanmartín C. Metal-based compounds containing selenium: An appealing approach towards novel therapeutic drugs with anticancer and antimicrobial effects. Eur J Med Chem 2022; 244:114834. [PMID: 36215861 DOI: 10.1016/j.ejmech.2022.114834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/22/2022] [Accepted: 10/02/2022] [Indexed: 11/17/2022]
|
32
|
Zhao R, Zhao H, Guo Q, Mu Y, Zhang J, Su Y, Han Q. Edaravone protects against liver fibrosis progression via decreasing the IL-1β secretion of macrophages. Chem Biol Interact 2022; 368:110251. [PMID: 36343683 DOI: 10.1016/j.cbi.2022.110251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/22/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
Edaravone (EDA), a strong novel free radical scavenger, have been demonstrated to exert neurovascular protective effects clinically. Furthermore, EDA can suppress the lung injury, pulmonary fibrosis and skin fibrosis, while the precise effects and mechanisms of EDA on liver injury and fibrosis remain unclear. The effects of EDA on the Thioacetamide (TAA)-induced liver fibrosis were evaluated by sirius red staining, α-SMA immunohistochemistry. The percentages of immune cell subsets were analyzed by flow cytometry. Immunofluorescence assay was performed to identify the fibrotic properties of hepatic stellate cells (HSCs). Western blot and qPCR were used to detect the levels of liver fibrosis-related molecules and IL-1β. EDA displayed a hepatic protective role in TAA-induced chronic liver fibrosis via inhibiting monocyte/macrophages recruitment and IL-1β production of macrophages. Mechanically, EDA inhibited of NF-κB signal pathway and reactive oxygen species (ROS) production in macrophages. Moreover, EDA treatment indirectly suppressed the activation of HSCs by decreasing the IL-1β secretion of macrophages. Together, EDA protects against TAA-induced liver fibrosis via decreasing the IL-1β production of macrophages, thereby providing a feasible solution for clinical treatment of liver fibrosis.
Collapse
Affiliation(s)
- Rongrong Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Quanjuan Guo
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Yongliang Mu
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Yuhang Su
- Department of Emergency Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
33
|
Zhan M, Guo Y, Shen M, Shi X. Nanomaterial‐Boosted Tumor Immunotherapy Through Natural Killer Cells. ADVANCED NANOBIOMED RESEARCH 2022; 2. [DOI: 10.1002/anbr.202200096] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
Natural killer (NK)‐cell immunotherapy as an alternative to T‐cell immunotherapy has been widely used in clinical cell immunotherapy of various tumors. Despite the surprising findings, the widespread applications of NK cells are still limited by the insufficient expansion and short lifespan of adoptive NK cells in vivo, the poor penetration of NK cells in solid tumors, as well as the immunosuppressive tumor microenvironment that may cause the inactivation of NK cells. Fortunately, the emergence of nanomaterials provides many opportunities to address these vexing problems, thus overcoming the barriers faced by NK cells and promoting the tumor inhibitory efficacy of NK cells. Herein, the recent advances in the rational design of nanomaterials for boosting the NK cell‐based immunotherapy, mainly through enhancing NK cell engagement with tumors, boosting NK cell activation or expansion, as well as redirecting NK cells to tumor cells, are reviewed. Lastly, the design and preparation of next‐generation nanomaterials that aim to further boost the NK cell‐based immunotherapy are briefly discussed.
Collapse
Affiliation(s)
- Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine College of Biological Science and Medical Engineering Donghua University Shanghai 201620 P.R. China
| |
Collapse
|
34
|
Du Y, Zhang Z, Yang Y, Liu T, Chen T, Li X. Highly active selenium nanotherapeutics combined with metformin to achieve synergistic sensitizing effect on NK cells for osteosarcoma therapy. NANOPHOTONICS (BERLIN, GERMANY) 2022; 11:5101-5111. [PMID: 39634308 PMCID: PMC11501141 DOI: 10.1515/nanoph-2022-0289] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/12/2022] [Indexed: 12/07/2024]
Abstract
NK cells-based cancer therapy combined with chemotherapeutic drugs for the treatment of tumors can enhance the immunosensitivity of NK cells, increase the expression of NK cell receptors, and eventually boost the killing effect of NK cells on cancer cells. Selenium (Se) with different chemical structures can be metabolized into selenoproteins to regulate tumor and immune cells' fate and functions. Herein, we found that, functionalized Se nanoparticles (SeNPs) combining with metformin (met) could amply the immunotherapeutic effects of NK92 cells against osteosarcoma cancer. The results revealed that TW80-SeNPs combined with met had the optimum performance on NK92 cells for HepG2 cells, owing to the increased ROS in HepG2 cells and the augmented expression of cell surface receptor proteins ULBP-3/4, PD-L1, MICA, and NK92 cell surface receptor proteins PD-1 and FasL. Additionally, TW80-SeNPs were gradually metabolized into selenoproteins (Gpx4 and TR1) into human osteosarcoma MG63 cells to reinforce the anticancer effect of NK92 cells by regulating the redox balance in the tumor microenvironment. This study provides a therapeutic approach in treating cancer itself or diabetes coupled with cancer. Moreover, it provides a multidrug strategy to improve immune cell function in practical applications, especially for synergistic immunotherapy of osteosarcoma.
Collapse
Affiliation(s)
- Yanxin Du
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Zehang Zhang
- Department of Oncology, Department of Chemistry, The First Affiliated Hospital, Jinan University, Guangzhou510632, China
| | - Yu Yang
- Department of Oncology, Department of Chemistry, The First Affiliated Hospital, Jinan University, Guangzhou510632, China
| | - Ting Liu
- Department of Oncology, Department of Chemistry, The First Affiliated Hospital, Jinan University, Guangzhou510632, China
| | - Tianfeng Chen
- Department of Oncology, Department of Chemistry, The First Affiliated Hospital, Jinan University, Guangzhou510632, China
| | - Xiaoling Li
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou510632, China
| |
Collapse
|
35
|
Lu Y, Zhu D, Le Q, Wang Y, Wang W. Ruthenium-based antitumor drugs and delivery systems from monotherapy to combination therapy. NANOSCALE 2022; 14:16339-16375. [PMID: 36341705 DOI: 10.1039/d2nr02994d] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Ruthenium complex is an important compound group for antitumor drug research and development. NAMI-A, KP1019, TLD1433 and other ruthenium complexes have entered clinical research. In recent years, the research on ruthenium antitumor drugs has not been limited to single chemotherapy drugs; other applications of ruthenium complexes have emerged such as in combination therapy. During the development of ruthenium complexes, drug delivery forms of ruthenium antitumor drugs have also evolved from single-molecule drugs to nanodrug delivery systems. The review summarizes the following aspects: (1) ruthenium complexes from monotherapy to combination therapy, including the development of single-molecule compounds, carrier nanomedicine, and self-assembly of carrier-free nanomedicine; (2) ruthenium complexes in the process of ADME in terms of absorption, distribution, metabolism and excretion; (3) the applications of ruthenium complexes in combination therapy, including photodynamic therapy (PDT), photothermal therapy (PTT), photoactivated chemotherapy (PACT), immunotherapy, and their combined application; (4) the future prospects of ruthenium-based antitumor drugs.
Collapse
Affiliation(s)
- Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
- Department of Chemistry, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway
| | - Di Zhu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
| | - Quynh Le
- Center for Pharmacy, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway.
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, P. R. China.
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing Laboratory of Oral Health, Beijing 100069, P. R. China
| | - Wei Wang
- Center for Pharmacy, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway.
- Department of Chemistry, University of Bergen, P. O. Box 7803, 5020 Bergen, Norway
| |
Collapse
|
36
|
Hu T, Gong H, Xu J, Huang Y, Wu F, He Z. Nanomedicines for Overcoming Cancer Drug Resistance. Pharmaceutics 2022; 14:pharmaceutics14081606. [PMID: 36015232 PMCID: PMC9412887 DOI: 10.3390/pharmaceutics14081606] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Clinically, cancer drug resistance to chemotherapy, targeted therapy or immunotherapy remains the main impediment towards curative cancer therapy, which leads directly to treatment failure along with extended hospital stays, increased medical costs and high mortality. Therefore, increasing attention has been paid to nanotechnology-based delivery systems for overcoming drug resistance in cancer. In this respect, novel tumor-targeting nanomedicines offer fairly effective therapeutic strategies for surmounting the various limitations of chemotherapy, targeted therapy and immunotherapy, enabling more precise cancer treatment, more convenient monitoring of treatment agents, as well as surmounting cancer drug resistance, including multidrug resistance (MDR). Nanotechnology-based delivery systems, including liposomes, polymer micelles, nanoparticles (NPs), and DNA nanostructures, enable a large number of properly designed therapeutic nanomedicines. In this paper, we review the different mechanisms of cancer drug resistance to chemotherapy, targeted therapy and immunotherapy, and discuss the latest developments in nanomedicines for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Hanlin Gong
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Jiayue Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Yuan Huang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
| | - Fengbo Wu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Correspondence: (F.W.); or (Z.H.); Tel.: +86-28-85422965 (Z.H.); Fax: +86-28-85422664 (Z.H.)
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China; (T.H.); (J.X.); (Y.H.)
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Correspondence: (F.W.); or (Z.H.); Tel.: +86-28-85422965 (Z.H.); Fax: +86-28-85422664 (Z.H.)
| |
Collapse
|
37
|
Chen G, Yang F, Fan S, Jin H, Liao K, Li X, Liu GB, Liang J, Zhang J, Xu JF, Pi J. Immunomodulatory roles of selenium nanoparticles: Novel arts for potential immunotherapy strategy development. Front Immunol 2022; 13:956181. [PMID: 35958612 PMCID: PMC9361286 DOI: 10.3389/fimmu.2022.956181] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/29/2022] [Indexed: 12/27/2022] Open
Abstract
Current chemotherapy strategies used in clinic appear with lots of disadvantages due to the low targeting effects of drugs and strong side effects, which significantly restricts the drug potency, causes multiple dysfunctions in the body, and even drives the emergence of diseases. Immunotherapy has been proved to boost the body’s innate and adaptive defenses for more effective disease control and treatment. As a trace element, selenium plays vital roles in human health by regulating the antioxidant defense, enzyme activity, and immune response through various specific pathways. Profiting from novel nanotechnology, selenium nanoparticles have been widely developed to reveal great potential in anticancer, antibacterial, and anti-inflammation treatments. More interestingly, increasing evidence has also shown that functional selenium nanoparticles can be applied for potential immunotherapy, which would achieve more effective treatment efficiency as adjunctive therapy strategies for the current chemotherapy. By directly interacting with innate immune cells, such as macrophages, dendritic cells, and natural killer cells, selenium nanoparticles can regulate innate immunity to intervene disease developments, which were reported to boost the anticancer, anti-infection, and anti-inflammation treatments. Moreover, selenium nanoparticles can also activate and recover different T cells for adaptive immunity regulations to enhance their cytotoxic to combat cancer cells, indicating the potential of selenium nanoparticles for potential immunotherapy strategy development. Here, aiming to enhance our understanding of the potential immunotherapy strategy development based on Se NPs, this review will summarize the immunological regulation effects of selenium nanoparticles and the application of selenium nanoparticle-based immunotherapy strategies. Furthermore, we will discuss the advancing perspective of selenium nanoparticle-based potential immunotherapy as a kind of novel adjunctive therapy to enhance the efficiency of current chemotherapies and also introduce the current obstacles for the development of selenium nanoparticles for potential immunotherapy strategy development. This work is expected to promote the future research on selenium nanoparticle-assisted immunotherapy and finally benefit the more effective disease treatments against the threatening cancer and infectious and chronic diseases.
Collapse
Affiliation(s)
- Gengshi Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Fen Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Shuhao Fan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Kangsheng Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xuemeng Li
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Pathogenic Biology and Immunology, School of Basic Medicine, Guangdong Medical University, Dongguan, China
| | - Gan-Bin Liu
- Department of Respiration, Dongguan 6th Hospital, Dongguan, China
| | - Jing Liang
- Department of Respiration, Dongguan 6th Hospital, Dongguan, China
| | - Junai Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Junai Zhang, ; Jun-Fa Xu, ; Jiang Pi,
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Junai Zhang, ; Jun-Fa Xu, ; Jiang Pi,
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
- *Correspondence: Junai Zhang, ; Jun-Fa Xu, ; Jiang Pi,
| |
Collapse
|
38
|
Wei Z, Yi Y, Luo Z, Gong X, Jiang Y, Hou D, Zhang L, Liu Z, Wang M, Wang J, Guo R, Yang J, Wang L, Wang H, Zhao Y. Selenopeptide Nanomedicine Activates Natural Killer Cells for Enhanced Tumor Chemoimmunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108167. [PMID: 35132688 DOI: 10.1002/adma.202108167] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/18/2022] [Indexed: 06/14/2023]
Abstract
Chemoimmunotherapy using nanotechnology has shown great potential for cancer therapy in the clinic. However, uncontrolled transportation and synergistic responses remain challenges. Here, a self-assembled selenopeptide nanoparticle that strengthens tumor chemoimmunotherapy through the activation of natural killer (NK) cells by the oxidative metabolite of the selenopeptide is developed. With the advantages of the enzyme-induced size-reduction and the reactive-oxygen-species-driven deselenization, this selenopeptide is able to deliver therapeutics, e.g., doxorubicin (DOX), to solid tumors and further activate the NK cells in a programmed manner. Importantly, in vitro and in vivo results prove the mutual promotion between the DOX-induced chemotherapy and the selenopeptide-induced immunotherapy, which synergistically contribute to the improved antitumor efficacy. It is anticipated that the selenopeptide may provide a type of promising stimuli-responsive immune modulator for versatile biomedical applications.
Collapse
Affiliation(s)
- Ziyu Wei
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin, 300384, China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhen Luo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, 18, Beisanhuandonglu, Chaoyang District, Beijing, 100029, China
| | - Yuxing Jiang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Dayong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Li Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Zimo Liu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Mandi Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Jie Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Ruochen Guo
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Jinjun Yang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin, 300384, China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Yuliang Zhao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| |
Collapse
|
39
|
Zheng N, Fu Y, Liu X, Zhang Z, Wang J, Mei Q, Wang X, Deng G, Lu J, Hu J. Tumor microenvironment responsive self-cascade catalysis for synergistic chemo/chemodynamic therapy by multifunctional biomimetic nanozymes. J Mater Chem B 2022; 10:637-645. [PMID: 34991154 DOI: 10.1039/d1tb01891d] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chemodynamic therapy (CDT) is an emerging approach to treat cancer based on the tumor microenvironment (TME), but its limited content of endogenous hydrogen peroxide (H2O2) weakens the anticancer effects. Herein, a multifunctional biomimetic nanozyme (Se@SiO2-Mn@Au/DOX, named as SSMA/DOX) is fabricated, which undergoes TME responsive self-cascade catalysis to facilitate MRI guided enhanced chemo/chemodynamic therapy. The SSMA/DOX nanocomposites (NCs) responsively degrade in acidic conditions of tumor to release Se, DOX, Au and Mn2+. Mn2+ not only enables MRI to guided therapy, but also catalyzes the endogenous H2O2 into hydroxyl radical (˙OH) for CDT. In addition, the Au NPs continuously catalyze glucose to generate H2O2, enhancing CDT by supplementing a sufficiently reactive material and cutting off the energy supply of the tumor by consuming glucose. Simultaneously, Se enhances the chemotherapy of doxorubicin hydrochloride (DOX) and CDT by upregulating ROS in the tumor cells, achieving remarkable inhibition effect towards tumor. Moreover, SSMA/DOX NCs have good biocompatibility and degradability, which avoid long-term toxicity and side effects. Overall, the degradable SSMA/DOX NCs provide an innovative strategy for tumor microenvironment responsive self-cascade catalysis to enhance tumor therapy.
Collapse
Affiliation(s)
- Nannan Zheng
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China. .,College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China.
| | - Yang Fu
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China.,Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China
| | - Xijian Liu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Ziwen Zhang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Jinxia Wang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Qixiang Mei
- Shanghai Key Laboratory of Pancreatic Disease, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China.,Department of Gastroenterology, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201600, China
| | - Xingyan Wang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 201620, China
| | - Jie Lu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| | - Junqing Hu
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen 518118, China. .,Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
40
|
Ruthenium complexes boost NK cell immunotherapy via sensitizing triple-negative breast cancer and shaping immuno-microenvironment. Biomaterials 2022; 281:121371. [DOI: 10.1016/j.biomaterials.2022.121371] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 01/03/2022] [Accepted: 01/08/2022] [Indexed: 11/23/2022]
|
41
|
Yang Y, Zhang Z, Chen Q, You Y, Li X, Chen T. Functionalized Selenium Nanoparticles Synergizes With Metformin to Treat Breast Cancer Cells Through Regulation of Selenoproteins. Front Bioeng Biotechnol 2021; 9:758482. [PMID: 34708029 PMCID: PMC8543061 DOI: 10.3389/fbioe.2021.758482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 08/26/2021] [Indexed: 12/19/2022] Open
Abstract
Owing to high blood sugar level and chronic inflammation, diabetes tend to cause the overproduction of free radicals in body, which will damage tissue and cells, reduce autoimmunity, and greatly increase the incidence of tumors. Selenium nanoparticles (SeNPs) exhibit high antioxidant activity with anti-tumor ability. In addition, metformin is considered as a clinical drug commonly for the treatment of stage II diabetes. Therefore, in this study, different functionalized SeNPs combined with metformin were performed to detect the feasibility for cancer therapy. The combination of Tween 80 (TW80)-SeNPs and metformin was found to have a synergistic effect on MCF-7 cells. The mechanism of this synergistic effect involved in the induction of DNA damage by affecting the generation of reactive oxygen species through selenoproteins; the upregulation of DNA-damage-related proteins including p-ATM, p-ATR, and p38; the promotion of p21 expression; and the downregulation of cyclin-dependent kinases and cyclin-related proteins causing cell cycle arrest. Furthermore, the expression of AMPK was affected, which in turn to regulate the mitochondrial membrane potential to achieve the synergistic treatment effect.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoling Li
- Department of Chemistry, and Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
| | - Tianfeng Chen
- Department of Chemistry, and Institute of Food Safety and Nutrition, Jinan University, Guangzhou, China
| |
Collapse
|
42
|
Selenium stimulates the antitumour immunity: Insights to future research. Eur J Cancer 2021; 155:256-267. [PMID: 34392068 DOI: 10.1016/j.ejca.2021.07.013] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 01/10/2023]
Abstract
Selenium is an essential trace element for regulating immune functions through redox-regulating activity of selenoproteins (e.g. glutathione peroxidase), protecting immune cells from oxidative stress. However, in cancer, selenium has biological bimodal action depending on the concentration. At nutritional low doses, selenium, depending on its form, may act as an antioxidant, protecting against oxidative stress, supporting cell survival and growth, thus, plays a chemo-preventive role; while, at supra-nutritional higher pharmacological doses, selenium acts as pro-oxidant inducing redox signalling and cell death. To date, many studies have been conducted on the benefits of selenium intake in reducing the risk of cancer incidence at the nutritional level, indicating that likely selenium functions as an immunostimulator, i.e. reversing the immunosuppression in tumour microenvironment towards antitumour immunity by activating immune cells (e.g. M1 macrophages and CD8+ T-lymphocytes) and releasing pro-inflammatory cytokines such as interferon-gamma; whereas, fewer studies have explored the effects of supra-nutritional or pharmacological doses of selenium in cancer immunity. This review, thus, systematically analyses the current knowledge about how selenium stimulates the immune system against cancer and lay the groundwork for future research. Such knowledge can be promising to design combinatorial therapies with Selenium-based compounds and other modalities like immunotherapy to lower the adverse effects and increase the efficacy of treatments.
Collapse
|
43
|
Song Z, Luo W, Zheng H, Zeng Y, Wang J, Chen T. Translational Nanotherapeutics Reprograms Immune Microenvironment in Malignant Pleural Effusion of Lung Adenocarcinoma. Adv Healthc Mater 2021; 10:e2100149. [PMID: 33870649 DOI: 10.1002/adhm.202100149] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/05/2021] [Indexed: 12/24/2022]
Abstract
Malignant pleural effusion (MPE) remains a treatment bottleneck in advanced lung cancer, due to its complicated microenvironments and "cold" immunity. Therefore, the search for therapeutic drugs to transform MPE to functionally "hot" one could advance the development of effective immunotherapeutic strategy. Herein, translational selenium nanoparticles coated with immune-modulating macromolecule lentinan (SeNPs@LNT) are designed to restore the dysfunctional immune cells in patient-derived MPE microenvironment. Internalization of the SeNPs@LNT can effectively reduce the immunosuppressive status by enhancing the proliferation of CD4+ T cells and natural killer cells, and remodeling the tumor associated macrophages into tumoricidal M1 phenotype in MPE derived from patients presenting low Se levels in blood and pleural effusion. Th1, cytotoxic T cell, γδ T, and B cell functions are upregulated, and Th2, Th17, and Treg cells activity is downregulated. Furthermore, SeNPs@LNT can be gradually metabolized into SeCys2 to promote the production of metabolites associated with tumor growth inhibition and immune response activation in MPE microenvironment. In contrast, lung cancer markers and vitamin B6 metabolism are decreased. The translational SeNP-based nanotherapeutic strategy restores functional "cold" MPE to "hot" MPE to activate the immune responses of various immune cells in MPE of lung cancer patients.
Collapse
Affiliation(s)
- Zhenhuan Song
- Research Center of Cancer Diagnosis and Therapy Department of Oncology The First Affiliated Hospital and Department of Chemistry Jinan University Guangzhou 510632 China
| | - Weizhan Luo
- Department of Respiratory Disease The State Key Laboratory of Respiratory Disease China Clinical Research Centre for Respiratory Disease Guangzhou Institute of Respiratory Disease First Affiliated Hospital of Guangzhou Medical University Guangzhou 510120 China
| | - Haichong Zheng
- Department of Respiratory Disease The State Key Laboratory of Respiratory Disease China Clinical Research Centre for Respiratory Disease Guangzhou Institute of Respiratory Disease First Affiliated Hospital of Guangzhou Medical University Guangzhou 510120 China
| | - Yunxiang Zeng
- Department of Respiratory Disease The State Key Laboratory of Respiratory Disease China Clinical Research Centre for Respiratory Disease Guangzhou Institute of Respiratory Disease First Affiliated Hospital of Guangzhou Medical University Guangzhou 510120 China
| | - Jinlin Wang
- Department of Respiratory Disease The State Key Laboratory of Respiratory Disease China Clinical Research Centre for Respiratory Disease Guangzhou Institute of Respiratory Disease First Affiliated Hospital of Guangzhou Medical University Guangzhou 510120 China
| | - Tianfeng Chen
- Research Center of Cancer Diagnosis and Therapy Department of Oncology The First Affiliated Hospital and Department of Chemistry Jinan University Guangzhou 510632 China
| |
Collapse
|
44
|
|
45
|
Huang X, Ou C, Shu Y, Wang Y, Gong S, Luo R, Chen S, Wu Q, Gong C. A self-sustained nanoplatform reverses TRAIL-resistance of pancreatic cancer through coactivating of exogenous and endogenous apoptotic pathway. Biomaterials 2021; 272:120795. [PMID: 33836292 DOI: 10.1016/j.biomaterials.2021.120795] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/27/2021] [Accepted: 03/29/2021] [Indexed: 02/05/2023]
Abstract
Since the 5-year survival rate of pancreatic cancer is only 10.0%, new therapies are urgently needed. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis specifically on tumor cells, nevertheless its clinical application was seriously restricted by resistance and short in vivo half-life. Herein, a novel multifunctional R6ST protein equipped with cell penetrating peptides R6, intrinsic apoptosis inducing tetrapeptide AVPI and soluble TRAIL was designed and constructed. Then, it was recruited to prepare self-sustained nanoplatform (SSN) to reverse TRAIL-resistance of pancreatic cancer through simultaneously promoting extrinsic and intrinsic apoptotic pathway, as well to elongate circulation time. Once administrated, high tumor accumulation and cellular uptake of SSN were achieved through prolonged circulation time, targeting ability of soluble TRAIL to death receptors and positive-charged R6, and further enhanced through reversed upregulation of death receptors on TRAIL-resistant tumor cells by the cumulated artesunate released in cytoplasm as a positive feedback loop. Furthermore, this loop simultaneously promoted extrinsic apoptosis of TRAIL fragment via the upregulated death receptors on TRAIL-resistant pancreatic cancer cells and intrinsic apoptosis of AVPI tetrapeptide via the efficient accumulation and uptake of R6ST on SSN. Hence, SSN exhibited synergistic antitumor effect and provided a new strategy for TRAIL-resistant pancreatic cancer therapy.
Collapse
Affiliation(s)
- Xianzhou Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chunqing Ou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaqian Shu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Songlin Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shouchun Chen
- Chengdu Huachuang Biotechnology Co. Ltd., Chengdu, 610041, China
| | - Qinjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
46
|
Chan L, Chen X, Gao P, Xie J, Zhang Z, Zhao J, Chen T. Coordination-Driven Enhancement of Radiosensitization by Black Phosphorus via Regulating Tumor Metabolism. ACS NANO 2021; 15:3047-3060. [PMID: 33507069 DOI: 10.1021/acsnano.0c09454] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Coordination-driven surface modification is an effective strategy to achieve nanosystem functionalization and improved physicochemical performance. Black phosphorus (BP)-based nanomaterials demonstrate great potential in cancer therapy, but their poor stability, low X-ray mass attenuation coefficient, and nonselectivity limit the application in radiotherapy. Herein, we used unsaturated iridium complex to coordinate with BP nanosheets to synthesize a two-dimensional layered nanosystem (RGD-Ir@BP) with higher biostability. Ir complex improves the photoelectric properties and photoinduced charge carrier dynamics of BP, hence Ir@BP generated more singlet oxygen after X-ray irradiation. In in vivo experiments, with X-ray irradiation, RGD-Ir@BP effectively inhibited nasopharyngeal carcinoma tumor growth but with minor side effects. Additionally, based on untargeted metabolomics analysis, the combined treatment specifically down-regulated the tumor proliferative mark of prostaglandin E2 in cancer cells. In general, this study provides a design strategy of high-performance coordination-driven BP-based nanosensitizer in cancer radiotherapy.
Collapse
Affiliation(s)
- Leung Chan
- Department of Oncology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, P. R. China
| | - Xiaodan Chen
- Department of Oncology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, P. R. China
| | - Pan Gao
- Department of Oncology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, P. R. China
| | - Jun Xie
- Department of Oncology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, P. R. China
| | - Zhongyang Zhang
- Department of Oncology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, P. R. China
| | - Jianfu Zhao
- Department of Oncology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, P. R. China
| | - Tianfeng Chen
- Department of Oncology, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
47
|
Wu YX, Lu HF, Lin YH, Chuang HY, Su SC, Liao YJ, Twu YC. Branched I antigen regulated cell susceptibility against natural killer cytotoxicity through its N-linked glycosylation and overall expression. Glycobiology 2021; 31:624-635. [PMID: 33403394 DOI: 10.1093/glycob/cwaa117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 12/16/2020] [Indexed: 11/14/2022] Open
Abstract
Cell surface glycosylation has been known as an important modification process that can be targeted and manipulated by malignant cells to escape from host immunosurveillance. We previously showed that the blood group branched I antigen on the leukemia cell surface can regulate the cell susceptibility against natural killer (NK) cell-mediated cytotoxicity through interfering target-NK interaction. In this work, we first identified N-linkage as the major glycosylation linkage type for branched I glycan formation on leukemia cells, and this linkage was responsible for cell sensitivity against therapeutic NK-92MI targeting. Secondly, by examining different leukemia cell surface death receptors, we showed death receptor Fas had highest expressions in both Raji and TF-1a cells. Mutations on two Fas extracellular N-linkage sites (118 and 136) for glycosylation impaired activation of Fas-mediated apoptosis during NK-92MI cytotoxicity. Last, we found that the surface I antigen expression levels enable leukemia cells to respond differently against NK-92MI targeting. In low I antigen expressing K-562 cell, reduction of I antigen presence greatly reduced leukemia cell susceptibility against NK-92MI targeting. But in other high I antigen expressing leukemia cells, similar reduction in I antigen expression did not affect cell susceptibility.
Collapse
Affiliation(s)
- Yu-Xuan Wu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, 155, Sec. 2, Li-Nong-St., Taipei, 112, Taiwan
| | - Hsu-Feng Lu
- Department of Clinical Pathology, Cheng Hsin General Hospital, 45, Cheng-Hsin St., Taipei, 112, Taiwan.,Department of Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, 510, Zhongzheng Rd., New Taipei City, 242, Taiwan
| | - Yen-Hsi Lin
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, 155, Sec. 2, Li-Nong-St., Taipei, 112, Taiwan.,Department of Clinical Laboratory, Chung Shan Medical University Hospital, 110, Sec. 1, Jianguo N. Rd., Taichung City, 402, Taiwan
| | - Hui-Yu Chuang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, 155, Sec. 2, Li-Nong-St., Taipei, 112, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, 222, Maijin Rd., Keelung City, 204, Taiwan.,Central Research Laboratory, Xiamen Chang Gung Hospital, 123, Xiafei Rd., Haicang District, Xiamen, China
| | - Yi-Jen Liao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, 250 Wu-Hsing St., Taipei, 110, Taiwan
| | - Yuh-Ching Twu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, 155, Sec. 2, Li-Nong-St., Taipei, 112, Taiwan
| |
Collapse
|
48
|
Han C, Wang Z, Xu Y, Chen S, Han Y, Li L, Wang M, Jin X. Roles of Reactive Oxygen Species in Biological Behaviors of Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1269624. [PMID: 33062666 PMCID: PMC7538255 DOI: 10.1155/2020/1269624] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa), known as a heterogenous disease, has a high incidence and mortality rate around the world and seriously threatens public health. As an inevitable by-product of cellular metabolism, reactive oxygen species (ROS) exhibit beneficial effects by regulating signaling cascades and homeostasis. More and more evidence highlights that PCa is closely associated with age, and high levels of ROS are driven through activation of several signaling pathways with age, which facilitate the initiation, development, and progression of PCa. Nevertheless, excessive amounts of ROS result in harmful effects, such as genotoxicity and cell death. On the other hand, PCa cells adaptively upregulate antioxidant genes to detoxify from ROS, suggesting that a subtle balance of intracellular ROS levels is required for cancer cell functions. The current review discusses the generation and biological roles of ROS in PCa and provides new strategies based on the regulation of ROS for the treatment of PCa.
Collapse
Affiliation(s)
- Chenglin Han
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zilong Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yingkun Xu
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Shuxiao Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yuqing Han
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Lin Li
- Department of Orthopedics, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Muwen Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xunbo Jin
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| |
Collapse
|
49
|
Liu C, Lai H, Chen T. Boosting Natural Killer Cell-Based Cancer Immunotherapy with Selenocystine/Transforming Growth Factor-Beta Inhibitor-Encapsulated Nanoemulsion. ACS NANO 2020; 14:11067-11082. [PMID: 32806028 DOI: 10.1021/acsnano.9b10103] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Natural killer (NK) cell-based immunotherapy represents a promising strategy to overcome the bottlenecks of cancer treatment. However, the therapeutic efficacy is greatly limited by downregulation of recognition ligands on the tumor cell surface, and the immunosuppressive effects can be thwarted by the tumor microenvironment such as secretion of transforming growth factor-beta (TGF-β), which could stunt the NK cell-mediated immune response. To overcome these limitations, herein we developed a nanoemulsion system (SSB NMs) to co-deliver TGF-β inhibitor and selenocysteine (SeC) to achieve amplified anticancer efficacy. SSB NMs significantly enhanced the lytic potency of NK92 cells by 2.1-fold. Moreover, a subtoxic dose of SSB NMs effectively sensitized MDA-MB-231 triple-negative breast cancer (TNBC) cells to NK cells derived from seven clinical patients, resulting in an up to 13.8-fold increase in cancer lysis. Mechanistic studies reveal that the sensitizing effects relied on natural killer group 2, member D (NKG2D)/NKG2D ligands (NKG2DLs) signaling with the involvement of DNA damage response. SSB NMs also effectively restrained TGF-β/TGF-β RI/Smad2/3 signaling, which thus enhanced NKG2DL expression on tumor cells and stimulated NKG2D surface expression on NK92 cells, ultimately contributing to the enhanced immune response. Furthermore, SSB NMs sustained release of SeC and TGF-β inhibitor and synergized with NK92 cells to induce significant anticancer effects in vivo. Together, this study not only demonstrates a simple strategy for the design of a nanoemulsion to co-deliver synergistic drugs but also sheds light on the application and action mechanisms in NK cell adaptive therapy against breast cancer, especially TNBCs.
Collapse
Affiliation(s)
- Chang Liu
- The First Affiliated Hospital and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Haoqiang Lai
- The First Affiliated Hospital and Department of Chemistry, Jinan University, Guangzhou 510632, China
| | - Tianfeng Chen
- The First Affiliated Hospital and Department of Chemistry, Jinan University, Guangzhou 510632, China
| |
Collapse
|
50
|
Li J, Chen T. Transition metal complexes as photosensitizers for integrated cancer theranostic applications. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213355] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|