1
|
Tao M, Li J, Zhang H, Zhan J, Wang X, Zhang K, Zhang J, Feng Z. Engineered L-phenylserine aldolase enhances L-norvaline synthesis within an enzyme cascade. J Biotechnol 2025; 405:99-110. [PMID: 40355092 DOI: 10.1016/j.jbiotec.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
L-Norvaline is a crucial intermediate in the synthesis of antihypertensive agents, and its production via biotechnological methods has garnered significant interest and commercial value in recent years. Here, for the enzymatic cascade synthesis of L-norvaline from propionaldehyde and glycine, an L-phenylserine aldolase gene (designated as ppLPA) from Pseudomonas putida was selected. Following the identification of potential mutation sites via error-prone PCR, coupled with site-directed mutagenesis, a single-site mutant, I18T, was identified, exhibiting a 1.4-fold increase in enzyme activity. Then, the ppLPA mutant I18T was combined with L-threonine deaminase, L-leucine dehydrogenase, and alcohol dehydrogenase to construct a one-pot, multi-enzyme cascade catalytic system for L-norvaline synthesis. The reaction conditions were systematically optimized. To mitigate the inhibitory effects of propionaldehyde, we employed a pH-stat substrate feeding strategy. Under optimal reaction conditions, L-norvaline production achieved a maximum yield of 116.5 g/L after 14 h of reaction, with a conversion rate exceeding 99 % in a 1 L reaction volume. This study highlights significant advancements in improving L-norvaline production, providing potential for more efficient biomanufacturing processes and broader industrial applications.
Collapse
Affiliation(s)
- Meijun Tao
- School of Life Science, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China
| | - Jing Li
- School of Life Science, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China
| | - Huaiyuan Zhang
- School of Agriculture, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China
| | - Jiuyu Zhan
- School of Life Science, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China
| | - Xinye Wang
- School of Life Science, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China
| | - Kai Zhang
- School of Life Science, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China
| | - Juan Zhang
- School of Agriculture, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China
| | - Zhibin Feng
- School of Life Science, Ludong University, 186 Hongqi Road, Yantai, Shandong 264025, China.
| |
Collapse
|
2
|
G H, Patil A, Mg H, Moqbel Redhwan MA, Guha S. "Development, optimization, and characterization of Eudragit-based nanoparticles for Dasatinib delivery". JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:756-778. [PMID: 39560680 DOI: 10.1080/09205063.2024.2427489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
This study focused on developing and evaluating dasatinib-loaded nanoparticles (DST-NPs) using Eudragit L100 as a polymer matrix for enhanced breast cancer treatment. The optimized formulation exhibited a particle size of 202.1 ± 5.7 nm, a zeta potential of -18 ± 1.01 mV, and an entrapment efficiency of 93.11 ± 0.2%. In-vitro release studies demonstrated sustained drug release from DST-NPs, following Fickian diffusion. Pharmacokinetic studies in rats revealed higher Cmax and AUC0-t for DST-NPs compared to pure DST, indicating improved bioavailability. Tissue distribution studies showed enhanced targeting of DST-NPs, with higher concentrations in the liver and spleen. In vivo efficacy in a DMBA-induced mammary carcinoma model demonstrated that DST-NPs significantly reduced tumor volume, maintained stable body weight, and improved survival rates compared to pure DST. Hematologic analysis indicated a favorable blood profile with DST-NPs, and histopathological examinations confirmed the restoration of normal mammary gland and liver architecture. MTT assays showed higher cytotoxicity of DST-NPs against MCF-7, MDA-MB231, and 4T1 cell lines, with lower IC50 values than pure DST. Stability studies indicated that DST-NPs maintained their properties over six months at various storage conditions. These findings highlight the potential of DST-NPs as an effective nanocarrier system for cancer therapy.
Collapse
Affiliation(s)
- Hemanth G
- Department of Pharmaceutics, KLE College of Pharmacy, Bengaluru, India
| | - Anasuya Patil
- Department of Pharmaceutics, KLE College of Pharmacy, Bengaluru, India
| | - Hariprasad Mg
- Department of Pharmacology, KLE College of Pharmacy, Bengaluru, India
| | | | - Sourav Guha
- Department of Pharmacology, KLE College of Pharmacy, Bengaluru, India
| |
Collapse
|
3
|
Lei D, Wang W, Zhao J, Zhou Y, Chen Y, Dai J, Qiu Y, Qi H, Li C, Liang B, Liu B, Wang Q, Li R. An injectable gambogic acid loaded nanocomposite hydrogel enhances antitumor effect by reshaping immunosuppressive tumor microenvironment. Mater Today Bio 2025; 31:101611. [PMID: 40104652 PMCID: PMC11919334 DOI: 10.1016/j.mtbio.2025.101611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
Gambogic acid(GA)is a natural compound that exhibits strong antitumor activity against a variety of tumors. However, its poor water solubility, low specificity, and high toxicity lead to inevitable systemic adverse effects. To minimize side effects, combining gambogic acid (GA) with delivery systems such as nanohydrogels to develop an in situ vaccine system (ISV) shows great promise. In this study, we loaded GA into a novel in situ nanocomposite hydrogel vaccine system (Gel-NPs@GA) along with a near-infrared (NIR) fluorescent dye, IR-1061. The Gel-NPs@GA system allowed for temperature-triggered gelation, simplifying injection and the in vivo formation of a drug-releasing gel, with near-infrared monitoring for drug metabolism. Slow, continuous release of gelatinase-targeted GA nanoparticles from the hydrogel occurs, followed by cleavage of mPEG-peptide-PCL conjugates by gelatinase, causing particle aggregation for endocytosis by tumor cells. This approach tackled solubility issues and curbs excessive GA release, boosting therapeutic drug levels. The sustained GA release induces tumor cell apoptosis, releasing tumor antigens and reprogramming the immune-suppressive tumor microenvironment. In the CT26 colorectal cancer mice model, this in situ vaccine system significantly inhibited tumor growth. By integrating information about immune cell clusters within the tumor microenvironment with RNA sequencing results, we hypothesized that Gel-NPs@GA could synergistically stimulate the immune response through various pathways, promote the maturation of dendritic cells (DCs), increase the infiltration of T cells, and thereby remodel the tumor's immune microenvironment.
Collapse
Affiliation(s)
- Dan Lei
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wanru Wang
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianhang Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Yingling Zhou
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Chen
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, Nanjing, China
| | - Juanjuan Dai
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuling Qiu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Haoyue Qi
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chunhua Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Baorui Liu
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| | - Qin Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| | - Rutian Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Clinical College of Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, China
| |
Collapse
|
4
|
Guo Y, Dong W, Sun D, Zhao X, Huang Z, Liu C, Sheng Y. Bacterial metabolites: Effects on the development of breast cancer and therapeutic efficacy (Review). Oncol Lett 2025; 29:210. [PMID: 40070782 PMCID: PMC11894516 DOI: 10.3892/ol.2025.14956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Evidence suggests that various gut metabolites significantly impact breast cancer (BC) and its treatment. However, the underlying mechanisms remain poorly understood and require further investigation. In the present study, the current literature was reviewed to evaluate the roles of microbial metabolites in the development of BC and its response to treatment. Microbial metabolites, including secondary bile acids, short-chain fatty acids, amino acid metabolites, lipopolysaccharide, nisin and pyocyanin, serve crucial roles in the BC microenvironment. Microbial metabolites promote BC invasion, metastasis and recurrence by facilitating cellular movement, epithelial-mesenchymal transition, cancer stem cell function and diapedesis. Furthermore, certain metabolites, such as trimethylamine N-oxide and L-norvaline, can alter the pharmacokinetics of chemotherapeutic drugs. The present review highlights the possible involvement of microbial metabolites and bacteriocins in BC carcinogenesis, development and metastasis. These metabolites could provide new insights for BC treatment strategies and are considered potential therapeutic targets.
Collapse
Affiliation(s)
- Yan Guo
- Department of Endocrinology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Wenyan Dong
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Dezheng Sun
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Xiang Zhao
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Zhiping Huang
- Department of Hepatobiliary Surgery and Organ Transplantation, General Hospital of Southern Theater Command of People's Liberation Army, Guangzhou, Guangdong 51000, P.R. China
| | - Chaoqian Liu
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yuan Sheng
- Department of Thyroid and Breast Surgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
5
|
Peng P, Cao J, Cheng W, Ming H, He B, Duan X, Li L, Tian Y, Nice EC, Zhang Z, Huang C, Zheng S. Manganese dioxide-based in situ vaccine boosts antitumor immunity via simultaneous activation of immunogenic cell death and the STING pathway. Acta Biomater 2025; 194:467-482. [PMID: 39832699 DOI: 10.1016/j.actbio.2025.01.029] [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: 09/28/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
In situ vaccine (ISV) can activate the anti-tumor immune system by inducing immunogenic cell death (ICD) at the tumor site. However, the development of tumor ISV still faces challenges due to insufficient tumor antigens released by tumor cells and the existence of tumor immunosuppressive microenvironment (TIME). Targeting the STING pathway has been reported to enhance the adjuvant effects of in situ tumor vaccines by initiating innate immunity. Based on this, we developed a potent in situ cancer vaccine, MBMA-RGD ISV, which simultaneously induces ICD and activates the STING pathway to achieve sustained anti-tumor immunity. Specifically, a water-soluble prodrug Mit-ALA was synthesized from the chemotherapeutic agent mitoxantrone (Mit) and the photosensitizer precursor 5-aminolevulinic acid (5-ALA) by pH-responsive ester bonds, which was then loaded into pre-synthesized BSA-MnO2 nanoparticles and functionalized with the targeting Arg-Gly-Asp (RGD) peptide to obtain MBMA-RGD ISV. This ISV actively targets tumor cells by binding integrin receptors and then gradually releases antitumor components in response to tumor microenvironment (TME). The released 5-ALA is metabolized in mitochondria to produce photosensitizer PpIX. Under laser irradiation, the photodynamic property of PpIX coupled with the photothermal effect of Mit synergistically induced ICD, resulting in the release of tumor antigens and evoking adaptive immunity. Meanwhile, released Mn2+ and Mit synergistically activate the STING pathway by inducing DNA damage, further enhancing antitumor immunity. Moreover, large amounts of oxygen released by MnO2 relieved the hypoxia microenvironment, thus sensitizing photodynamic therapy and improving the immunosuppressive state of TME. Therefore, MBMA-RGD ISV efficiently activates systemic antitumor immunity in vitro and in vivo, providing new strategies and ideas for the development of tumor ISV. STATEMENT OF SIGNIFICANCE: Using a biocompatible BSA-MnO2 nanoplatform, we developed a dual-prodrug tumor in situ vaccine (ISV) with tumor microenvironment-responsive action for synergistic cancer immunotherapy. Once internalized by tumor cells, the MBMA-RGD ISV responded to intracellular H+, H2O2, and GSH, releasing its therapeutic "cargo." Under laser irradiation, the combined effects of photodynamic therapy (PDT) and photothermal therapy (PTT) induced immunogenic cell death (ICD), effectively recruiting and stimulating dendritic cells (DCs). Concurrently, STING pathway activation, triggered by DNA damage, enhanced DC maturation. Moreover, the MnO2 component alleviated hypoxia within the tumor microenvironment by releasing significant amounts of oxygen, which facilitated the repolarization of macrophages from the M2 phenotype to the M1 phenotype. Therefore, MBMA-RGD ISV demonstrated potent suppression of tumor metastasis and recurrence without notable side effects in mouse tumor models.
Collapse
Affiliation(s)
- Peilan Peng
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Jiangjun Cao
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Wenting Cheng
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Bo He
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Xirui Duan
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200025, China
| | - Lei Li
- Department of anorectal surgery, Hospital of Chengdu University of Traditional Chinese Medicine and Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuan Tian
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Zhiqi Zhang
- Department of General Surgery, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, PR China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China.
| | - Shaojiang Zheng
- Hainan Cancer Center and Tumor Institute, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China.
| |
Collapse
|
6
|
Jin K, Chu X, Qian J. Arginine and colorectal cancer: Exploring arginine-related therapeutic strategies and novel insights into cancer immunotherapies. Int Immunopharmacol 2025; 148:114146. [PMID: 39879835 DOI: 10.1016/j.intimp.2025.114146] [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: 05/05/2024] [Revised: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Concerning the progression of societies and the evolution of lifestyle and dietary habits, the potential for the development of human malignancies, particularly colorectal cancer (CRC), has markedly escalated, positioning it as one of the most prevalent and lethal forms of cancer globally. Empirical evidence indicates that the metabolic processes of cancerous and healthy cells can significantly impact immune responses and the fate of tumors. Arginine, a multifaceted amino acid, assumes a crucial and paradoxical role in various metabolic pathways, as certain tumors exhibit arginine auxotrophy while others do not. Notably, CRC is classified as arginine non-auxotrophic, possessing the ability to synthesize arginine from citrulline. Systemic arginine deprivation and the inhibition of arginine uptake represent two prevalent therapeutic strategies in oncological treatment. However, given the divergent behaviors of tumors concerning the metabolism and synthesis of arginine, one of these therapeutic approaches-namely systemic arginine deprivation-does not apply to CRC. This review elucidates the characteristics of arginine uptake inhibition and systemic arginine deprivation alongside their respective benefits and limitations in CRC. Furthermore, the involvement of arginine in immunotherapeutic strategies is examined in light of the most recent discoveries on various human malignancies.
Collapse
Affiliation(s)
- Ketao Jin
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang 310003, China.
| | - Xiufeng Chu
- Department of General Surgery, Shaoxing Central Hospital, Shaoxing, Zhejiang 312030, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People's Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang 312500, China.
| |
Collapse
|
7
|
Ma M, Zhang Y, Pu K, Tang W. Nanomaterial-enabled metabolic reprogramming strategies for boosting antitumor immunity. Chem Soc Rev 2025; 54:653-714. [PMID: 39620588 DOI: 10.1039/d4cs00679h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Immunotherapy has become a crucial strategy in cancer treatment, but its effectiveness is often constrained. Most cancer immunotherapies focus on stimulating T-cell-mediated immunity by driving the cancer-immunity cycle, which includes tumor antigen release, antigen presentation, T cell activation, infiltration, and tumor cell killing. However, metabolism reprogramming in the tumor microenvironment (TME) supports the viability of cancer cells and inhibits the function of immune cells within this cycle, presenting clinical challenges. The distinct metabolic needs of tumor cells and immune cells require precise and selective metabolic interventions to maximize therapeutic outcomes while minimizing adverse effects. Recent advances in nanotherapeutics offer a promising approach to target tumor metabolism reprogramming and enhance the cancer-immunity cycle through tailored metabolic modulation. In this review, we explore cutting-edge nanomaterial strategies for modulating tumor metabolism to improve therapeutic outcomes. We review the design principles of nanoplatforms for immunometabolic modulation, key metabolic pathways and their regulation, recent advances in targeting these pathways for the cancer-immunity cycle enhancement, and future prospects for next-generation metabolic nanomodulators in cancer immunotherapy. We expect that emerging immunometabolic modulatory nanotechnology will establish a new frontier in cancer immunotherapy in the near future.
Collapse
Affiliation(s)
- Muye Ma
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Dr 2, Singapore, 117545, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Dr, Singapore, 117597, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Wei Tang
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
- Department of Pharmacy and Pharmaceutic Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| |
Collapse
|
8
|
Amaro-da-Cruz A, Rubio-Tomás T, Álvarez-Mercado AI. Specific microbiome patterns and their association with breast cancer: the intestinal microbiota as a potential biomarker and therapeutic strategy. Clin Transl Oncol 2025; 27:15-41. [PMID: 38890244 PMCID: PMC11735593 DOI: 10.1007/s12094-024-03554-w] [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: 03/13/2024] [Accepted: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Breast cancer (BC) is one of the most diagnosed cancers in women. Based on histological characteristics, they are classified as non-invasive, or in situ (tumors located within the milk ducts or milk lobules) and invasive. BC may develop from in situ carcinomas over time. Determining prognosis and predicting response to treatment are essential tools to manage this disease and reduce its incidence and mortality, as well as to promote personalized therapy for patients. However, over half of the cases are not associated with known risk factors. In addition, some patients develop resistance to treatment and relapse. Therefore, it is necessary to identify new biomarkers and treatment strategies that improve existing therapies. In this regard, the role of the microbiome is being researched as it could play a role in carcinogenesis and the efficacy of BC therapies. This review aims to describe specific microbiome patterns associated with BC. For this, a literature search was carried out in PubMed database using the MeSH terms "Breast Neoplasms" and "Gastrointestinal Microbiome", including 29 publications. Most of the studies have focused on characterizing the gut or breast tissue microbiome of the patients. Likewise, studies in animal models and in vitro that investigated the impact of gut microbiota (GM) on BC treatments and the effects of the microbiome on tumor cells were included. Based on the results of the included articles, BC could be associated with an imbalance in the GM. This imbalance varied depending on molecular type, stage and grade of cancer, menopause, menarche, body mass index, and physical activity. However, a specific microbial profile could not be identified as a biomarker. On the other hand, some studies suggest that the GM may influence the efficacy of BC therapies. In addition, some microorganisms and bacterial metabolites could improve the effects of therapies or influence tumor development.
Collapse
Affiliation(s)
- Alba Amaro-da-Cruz
- Department of Chemical Engineering, Faculty of Science, University of Granada, 18071, Granada, Spain
| | - Teresa Rubio-Tomás
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Ana I Álvarez-Mercado
- Instituto de Investigación Biosanitaria ibs.GRANADA, Complejo Hospitalario Universitario de Granada, 18014, Granada, Spain.
- Institute of Nutrition and Food Technology, Biomedical Research Center, University of Granada, 18016, Armilla, Spain.
- Department of Pharmacology School of Pharmacy, University of Granada, 18071, Granada, Spain.
| |
Collapse
|
9
|
Gao Q, He T, Chen L, Zhu S, Li C, Zeng Y, Luo S, Chen S, Chen X, Yu S, Ye Z, Wu ZS. Triangle-toothed gear occlude-guided universal nanotechnology constructs 3D symmetric DNA polyhedra with high assembly efficiency for precision cancer therapy. J Colloid Interface Sci 2025; 677:1045-1060. [PMID: 39178668 DOI: 10.1016/j.jcis.2024.08.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 08/04/2024] [Accepted: 08/12/2024] [Indexed: 08/26/2024]
Abstract
Chemotherapy is commonly used to treat malignant tumors. However, conventional chemotherapeutic drugs often cannot distinguish between tumor and healthy cells, resulting in adverse effects and reduced therapeutic efficacy. Therefore, zigzag-shaped gear-occlude-guided cymbal-closing (ZGC) DNA nanotechnology was developed based on the mirror-symmetry principle to efficiently construct symmetric DNA polyhedra. This nanotechnology employed simple mixing steps for efficient sequence design and assembly. A targeting aptamer was installed at a user-defined position using an octahedron as a model structure. Chemotherapeutic drug-loaded polyhedral objects were subsequently delivered into tumor cells. Furthermore, anticancer drug-loaded DNA octahedra were intravenously injected into a HeLa tumor-bearing mouse model. Assembly efficiency was almost 100 %, with no residual building blocks identified. Moreover, this nanotechnology required a few DNA oligonucleotides, even for complex polyhedrons. Symmetric DNA polyhedrons retained their structural integrity for 24 h in complex biological environments, guaranteeing prolonged circulation without drug leakage in the bloodstream and promoting efficient accumulation in tumor tissues. In addition, DNA octahedra were cleared relatively slowly from tumor tissues. Similarly, tumor growth was significantly inhibited in vivo, and a therapeutic outcome comparable to that of conventional gene-chemo combination therapy was observed. Moreover, no systemic toxicity was detected. These findings indicate the potential application of ZGC DNA nanotechnology in precision medicine.
Collapse
Affiliation(s)
- Qian Gao
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Tenghang He
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Linhuan Chen
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Shidan Zhu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Congcong Li
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Yi Zeng
- Department of Gastric Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), China
| | - Shasha Luo
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Shu Chen
- Department of Gastric Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), China
| | - Xiangru Chen
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China; Department of Gastric Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), China
| | - Suhong Yu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China.
| | - Zaisheng Ye
- Department of Gastric Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China.
| |
Collapse
|
10
|
Liu Y, Liu F, Zeng Y, Lin L, Yu H, Zhang S, Yang W. Hydrogel systems for spatiotemporal controlled delivery of immunomodulators: engineering the tumor immune microenvironment for enhanced cancer immunotherapy. Front Cell Dev Biol 2024; 12:1514595. [PMID: 39735340 PMCID: PMC11681625 DOI: 10.3389/fcell.2024.1514595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Tumor immunotherapy, modulating innate and adaptive immunity, has become an important therapeutic strategy. However, the tumor immune microenvironment's (TIME) complexity and heterogeneity challenge tumor immunotherapy. Hydrogel is a hydrophilic three-dimensional (3D) mesh structure with good biocompatibility and drug release control, which is widely used in drug delivery, agriculture, industry, etc. Hydrogels loaded with immune cells, cytokines, immune checkpoint inhibitors, and anti-tumor drugs can achieve targeted delivery and ultimately activate the immune response in the TIME. In this review, we will summarize the components of the TIME and their immune effects, the emerging immunomodulatory agents, the characteristics and functions of hydrogels, and how hydrogels regulate innate and adaptive immune cells in the TIME.
Collapse
Affiliation(s)
- Yanting Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Fang Liu
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
- College of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yan Zeng
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Liangbin Lin
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Hui Yu
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Sunfu Zhang
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Wenyong Yang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| |
Collapse
|
11
|
Peng N, Du Y, Liu J, Li D, Li Y, Deng K, Li L, Jia P, Min J, Lin J. Injectable Polydopamine Nanoparticle-Incorporated Hydrogels for Antiangiogenesis and Stimulating Tumoricidal Immunity to Inhibit Metastasis and Recurrence Postresection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64447-64462. [PMID: 39540317 DOI: 10.1021/acsami.4c10363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Surgical resection is still the main means for clinical treatments of breast cancer, but the postoperative immunosuppressive microenvironment and neoangiogenesis of the residual tumors easily lead to tumor metastasis and recurrence, which will further endanger patients' lives. The combination of antiangiogenic therapy and immunotherapy may promote the mutually reinforced cycle of immune reprogramming and vascular normalization to avoid tumor metastasis and recurrence. Herein, we prepared polydopamine nanoparticles for improving tissue adhesion and enriching tumor-associated antigens. This nanoregulator together with regorafenib (REG) was further incorporated into a hydrogel developed from grafting adipic acid dihydrazide onto 2,2,6,6-tetramethylpiperidine-1-oxyl radical oxidized chitin and oxidized hyaluronic acid, which was injectable at the cavity after subcutaneous tumor surgery with good mechanical properties and degradability. The system showed long-term release of REG. After combining with anti-PD-L1, the hydrogel applied to the surgical wound exhibited a reduction in tumor metastasis and recurrence. This effect was achieved by suppressing angiogenesis and enhancing antitumor immunity, characterized by increased levels of effector T lymphocytes and activation of dendritic cells within tumors, spleens, and draining lymph nodes. The injectable hydrogel offers a promising strategy for postoperative management aimed at preventing tumor metastasis and recurrence.
Collapse
Affiliation(s)
- Na Peng
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Yijing Du
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Jinhong Liu
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Danyang Li
- College of Textiles and Clothing, Yancheng Institute of Technology, Yancheng 224051, Jiangsu, P. R. China
| | - Yonghao Li
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Kai Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, P. R. China
| | - Lewei Li
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Peizhi Jia
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Juan Min
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, P. R. China
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| |
Collapse
|
12
|
Tan T, Chang W, Wang TL, Chen W, Chen X, Yang C, Yang D. pH-Responsive Charge-Reversal Smart Nanoparticles for Co-Delivery of Mitoxantrone and Copper Ions to Enhance Breast Cancer Chemo-Chemodynamic Combination Therapy. Int J Nanomedicine 2024; 19:11445-11462. [PMID: 39530107 PMCID: PMC11552415 DOI: 10.2147/ijn.s479125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose The poor delivery and limited penetration of nanoparticles into breast cancer tumors remain essential challenges for effective anticancer therapy. This study aimed to design a promising nanoplatform with efficient tumor targeting and penetration capability for effective breast cancer therapy. Methods A pH-sensitive mitoxantrone (MTO) and copper ion-loaded nanosystem functionalized with cyclic CRGDfK and r9 peptide (TPRN-CM) was rationally designed for chemo-chemodynamic combination therapy. TPRN-CM would be quiescent in blood circulation with the CRGDfK peptide on the surface of the nanoparticle to improve its targeting to the tumor. Then, the structure of TPRN-CM changes in the acidic tumor microenvironment, and the r9 peptide can be exposed to make a surface charge reversal to promote deep penetration in the tumor and facilitate their internalization by cancer cells, which was characterized using transmission electron microscopy, dynamic light scattering, flame atomic absorption, etc. The drug release behavior, anti-tumor effects in vivo and in vitro, and the biosafety of the nanoplatform were evaluated. Results TPRN-CM exhibited remarkable capability to load MTO and Cu2+ with good stability in serum. It can achieve pH-responsive charge reversal, MTO, and Cu2+ release, and can further generate toxic hydroxyl radicals in the presence of glutathione (GSH) and H2O2. In vitro experiments demonstrated that this nanoplatform significantly inhibited proliferation, migration, invasion activities and 3D-tumorsphere growth. In vivo experiments suggested that rationally designed TPRN-CM can be effectively delivered to breast cancer tumors with deep tumor penetration, thereby resulting in a notable reduction in tumor growth and suppression of lung metastasis without causing any apparent side effects. Conclusion The constructed TPRN-CM nanoplatform integrated tumor targeting, tumor penetration, drug-responsive release, and chemo-chemodynamic combination therapy, thereby providing an intelligent drug delivery strategy to improve the efficacy of breast cancer treatment.
Collapse
Affiliation(s)
- Tao Tan
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Weiyi Chang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
- College of Life Science, Jilin University, Changchun, 130012, People’s Republic of China
| | - Tian Long Wang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Wei Chen
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Xiaobing Chen
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Chunmiao Yang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| | - Dongsheng Yang
- College of Life Science, Zhuhai College of Science and Technology, Zhuhai, 519041, People’s Republic of China
| |
Collapse
|
13
|
Khaliq NU, Lee J, Kim Y, Kim J, Kim T, Yu S, Seo D, Sung D, Kim H. Tumor cell loaded thermosensitive hydrogel for photodynamic therapy associated tumor antigens release. Biochim Biophys Acta Gen Subj 2024; 1868:130703. [PMID: 39163944 DOI: 10.1016/j.bbagen.2024.130703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 08/22/2024]
Abstract
BACKGROUND Immunotherapy is a powerful strategy for treating cancer and can be used to inhibit the post-surgical relapse of tumors. METHODS To achieve this, a Cell@hydrogel was developed as a template using a mixture of CT26 tumor cells and Pluronic® F-127/gelatin. RESULTS The proposed mixture has a solution-to-gelation functionality and vice versa. The morphology of the Cell@hydrogel was characterized by scanning electron microscopy and confocal microscopy. For photodynamic immunotherapy, the Cell@hydrogel was functionalized with Cy7 (Cy7-Cell@hydrogel) to quantify reactive oxygen species in CT26 tumor cells. Gel electrophoresis and membrane integrity tests were performed to determine the efficiency of the Cy7-Cell@hydrogel following photodynamic therapy. CONCLUSIONS This protocol provides an alternative approach that mechanistically inhibits the post-surgical relapse of solid tumors based on immunotherapy.
Collapse
Affiliation(s)
- Nisar Ul Khaliq
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea.
| | - Juyeon Lee
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Yejin Kim
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Joohyeon Kim
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Taeho Kim
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea
| | - Sohyeon Yu
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea; Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Dongseong Seo
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea; Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Daekyung Sung
- Center for Bio-Healthcare Materials, Bio-Convergence Materials R&D Division, Korea Institute of Ceramic Engineering and Technology, 202 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju 28160, Republic of Korea.
| | - Hyungjun Kim
- Department of Chemistry and Bioscience, Kumoh National Institute of Technology, 61 Daehak-ro, Gumi 39177, Republic of Korea.
| |
Collapse
|
14
|
Li F, Li Z, Wei C, Xu L, Liang Y, Yan J, Li Y, He B, Sun C. Application of hydrogels for targeting cancer stem cells in cancer treatment. Biomed Pharmacother 2024; 180:117486. [PMID: 39321506 DOI: 10.1016/j.biopha.2024.117486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/28/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024] Open
Abstract
Cancer stem cells (CSCs) are a major hindrance to clinical cancer treatment. Owing to their high tumorigenic and metastatic potential, CSCs are vital in malignant tumor initiation, growth, metastasis, and therapeutic resistance, leading to tumorigenesis and recurrence. Compared with normal tumor cells, CSCs express high levels of surface markers (CD44, CD90, CD133, etc.) and activate specific signaling pathways (Wnt/β-catenin, Notch, and Hedgehog). Although Current drug delivery systems (DDS) precisely target CSCs, the heterogeneity and multidrug resistance of CSCs impede CSC isolation and screening. Conversely, hydrogel DDSs exhibit good biocompatibility and high drug delivery efficiency. Hydrogels are three-dimensional (3D) spatial structures for drug encapsulation that facilitate the controlled release of bioactive molecules. Hence, hydrogels can be loaded with drugs to precisely target CSCs. Their 3D structure can also culture non-CSCs and facilitate their transformation into CSCs. for identification and isolation. Given that their elastic modulus and stiffness characteristics reflect those of the cellular microenvironment, hydrogels can simulate extracellular matrix pathways and markers to regulate CSCs, disrupting the equilibrium between CSC and non-CSC transformation. This article reviews the CSC microenvironment, metabolism, signaling pathway, and surface markers. Additionally, we summarize the existing CSC targeting strategies and explore the application of hydrogels for CSC screening and treatment. Finally, we discuss potential advances in CSC research that may lead to curative measures for tumors through targeted and precise attacks on CSCs.
Collapse
Affiliation(s)
- Fashun Li
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China; Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Chen Wei
- Department of Pharmacy, Qingdao Women and Children's Hospital, Qingdao 266034, China
| | - Long Xu
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo 315211, China.
| | - Yan Liang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China.
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Yifei Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266073, China
| | - Bin He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Chong Sun
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
15
|
Kaur H, Gogoi B, Sharma I, Das DK, Azad MA, Pramanik DD, Pramanik A. Hydrogels as a Potential Biomaterial for Multimodal Therapeutic Applications. Mol Pharm 2024; 21:4827-4848. [PMID: 39290162 PMCID: PMC11462506 DOI: 10.1021/acs.molpharmaceut.4c00595] [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: 05/30/2024] [Revised: 09/07/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Hydrogels, composed of hydrophilic polymer networks, have emerged as versatile materials in biomedical applications due to their high water content, biocompatibility, and tunable properties. They mimic natural tissue environments, enhancing cell viability and function. Hydrogels' tunable physical properties allow for tailored antibacterial biomaterial, wound dressings, cancer treatment, and tissue engineering scaffolds. Their ability to respond to physiological stimuli enables the controlled release of therapeutics, while their porous structure supports nutrient diffusion and waste removal, fostering tissue regeneration and repair. In wound healing, hydrogels provide a moist environment, promote cell migration, and deliver bioactive agents and antibiotics, enhancing the healing process. For cancer therapy, they offer localized drug delivery systems that target tumors, minimizing systemic toxicity and improving therapeutic efficacy. Ocular therapy benefits from hydrogels' capacity to form contact lenses and drug delivery systems that maintain prolonged contact with the eye surface, improving treatment outcomes for various eye diseases. In mucosal delivery, hydrogels facilitate the administration of therapeutics across mucosal barriers, ensuring sustained release and the improved bioavailability of drugs. Tissue regeneration sees hydrogels as scaffolds that mimic the extracellular matrix, supporting cell growth and differentiation for repairing damaged tissues. Similarly, in bone regeneration, hydrogels loaded with growth factors and stem cells promote osteogenesis and accelerate bone healing. This article highlights some of the recent advances in the use of hydrogels for various biomedical applications, driven by their ability to be engineered for specific therapeutic needs and their interactive properties with biological tissues.
Collapse
Affiliation(s)
- Harpreet Kaur
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | - Bishmita Gogoi
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | - Ira Sharma
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | - Deepak Kumar Das
- Department
of Chemistry and Nanoscience, GLA University, Mathura, Uttar Pradesh 281 406, India
| | - Mohd Ashif Azad
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
| | | | - Arindam Pramanik
- Amity
Institute of Biotechnology, Amity University, Noida 201301, India
- School
of Medicine, University of Leeds, Leeds LS97TF, United Kingdom
| |
Collapse
|
16
|
Jin Q, Zhou X, Niu X, Ping C, Dong X, Duan D, Wang R, Chen Y, Pan F, Yang F, Yang X, Zhang G, Wang R, Zhang S, Ren G. Co-delivery of doxorubicin-dihydroartemisinin prodrug/TEPP-46 nano-liposomes for improving antitumor and decreasing cardiotoxicity in B16-F10 tumor-bearing mice. Colloids Surf B Biointerfaces 2024; 241:113992. [PMID: 38833960 DOI: 10.1016/j.colsurfb.2024.113992] [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: 03/16/2024] [Revised: 05/12/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024]
Abstract
In order to reduce the cardiotoxicity of doxorubicin (DOX) and improve its antitumor effect, dihydroartemisinin (DHA) and DOX prodrug (DOX-S-DHA) synthesized via a single sulfur bond was used with TEPP-46 to prepare nano-liposomes (DOX-S-DHA@TEPP-46 Lips). In which, TEPP-46 was expected to exert p53 bidirectional regulation to promote the synergistic antitumor effect of DOX and DHA while reducing cardiotoxicity. DOX-S-DHA@TEPP-46 Lips exhibited uniform particle size, good stability, and excellent redox-responsive activity. DOX-S-DHA@TEPP-46 Lips could significantly inhibit the proliferation of tumor cells, but had less cytotoxicity on normal cells. The presence of TEPP-46 increased the content of p53 protein, which further induced tumor cell apoptosis. DOX-S-DHA@TEPP-46 Lips had satisfactory long circulation to enhance the antitumor efficacy and reversed the cardiotoxicity of DOX in B16-F10 tumor-bearing mice. In conclusion, DOX-S-DHA@TEPP-46 Lips provides a new insight on creating sophisticated redox-sensitive nano-liposomes for cancer therapy as well as the decreased cardiotoxicity of DOX.
Collapse
Affiliation(s)
- Qiuyue Jin
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaohui Zhou
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaomin Niu
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Canqi Ping
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaozhou Dong
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Danyu Duan
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Rongrong Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Yi Chen
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Fei Pan
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Fan Yang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Xihua Yang
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Guoshun Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Ruili Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| | - Guolian Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan 030001, China; Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
17
|
Ilhami FB, Munasir, Gultom NS, Cheng CC. Zinc Oxide/Carbon Material-Embedded Supramolecular Drug Delivery System with Photoswitching Properties for Highly Selective and Effective Chemotherapy. ACS APPLIED BIO MATERIALS 2024; 7:5506-5518. [PMID: 38979905 DOI: 10.1021/acsabm.4c00638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Phototherapy has become a hopeful procedure for the treatment of cancer. Nevertheless, the straightforward creation of a theranostic system that can achieve both tumor localization and production of oxygen species is greatly desired yet remains a challenging endeavor. In this study, we synthesized spherical nanostructures by decorating zinc oxide (ZnO) with peanut shell-based carbon (PNS-C) in an aqueous solution. The PNS-C-decorated ZnO (ZnO/PNS-C)-embedded supramolecular system exhibited spontaneous self-assembly. The nanogels that are produced have several desirable characteristics, including exceptional resistance to degradation by light, highly stable nanostructures that form spontaneously in biological environments, outstanding ability to prevent the destruction of red blood cells, and a high level of sensitivity to changes in pH and light. Under light irradiation, the addition of ZnO/PNS-C-incorporated supramolecular provided high reactive oxygen species production. Moreover, in vitro cellular assays demonstrated ZnO/PNS-C-incorporated supramolecular exhibited highly selective and induced phototoxicity into cancer cells and no effect on the viability of normal cells both before and after irradiation. Overall, the ZnO/PNS-C-incorporated supramolecular system has the potential to stimulate advancements in phototherapy by utilizing highly tumor-selective therapeutic molecules. This can lead to a more effective targeted therapy for cancers.
Collapse
Affiliation(s)
- Fasih Bintang Ilhami
- Department of Natural Science, Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya, Surabaya 60231, Indonesia
| | - Munasir
- Department of Physics, Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya, Surabaya 60231, Indonesia
| | | | - Chih-Chia Cheng
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
- Advanced Membrane Materials Research Center, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| |
Collapse
|
18
|
Park JK, Piao Z, Lee HJ, Jeong B. Poly(l-threonine- co-l-threonine Succinate) Thermogels for Sustained Release of Lixisenatide. Biomacromolecules 2024; 25:4946-4955. [PMID: 38949062 DOI: 10.1021/acs.biomac.4c00376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Negatively charged poly(l-Thr-co-l-Thr succinate) (PTTs) was developed as a new thermogel. Aqueous PTT solutions underwent thermogelation over a concentration range of 6.0-8.3 wt %. Dynamic light scattering, FTIR, 1H NMR, and COSY spectra revealed the partial strengthening of the β-sheet conformation and the dehydration of PTTs during the transition. Extendin-4 was released from the PTTs thermogel with a large initial burst release, whereas positively charged lixisenatide significantly reduced its initial burst release to 25%, and up to 77% of the dose was released from the gel over 14 days. In vivo study revealed a high plasma concentration of lixisenatide over 5 days and hypoglycemic efficacy was observed for type II diabetic rats over 7-10 days. The biocompatible PTTs were degraded by subcutaneous enzymes. This study thus demonstrates an effective strategy for reducing the initial burst release of protein drugs from thermogels with the introduction of electrostatic interactions between the drug and the thermogel.
Collapse
Affiliation(s)
- Jin Kyung Park
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Zhengyu Piao
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Hyun Jung Lee
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| | - Byeongmoon Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, 52 Ewhayeodae-gil, Seodaemun-gu, Seoul 03760, Korea
| |
Collapse
|
19
|
Mondal J, Chakraborty K, Bunggulawa EJ, An JM, Revuri V, Nurunnabi M, Lee YK. Recent advancements of hydrogels in immunotherapy: Breast cancer treatment. J Control Release 2024; 372:1-30. [PMID: 38849092 DOI: 10.1016/j.jconrel.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/21/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
Breast cancer is the most prevalent cancer among women and the leading cause of cancer-related deaths in this population. Recent advances in Immunotherapy, or combined immunotherapy, offering a more targeted and less toxic approach, expand the survival rate of patients more than conventional treatment. Notably, hydrogels, a versatile platform provided promising avenues to combat breast cancer in preclinical studies and extended to clinical practices. With advantages such as the alternation of tumor microenvironment, immunomodulation, targeted delivery of therapeutic agents, and their sustained release at specific sites of interest, hydrogels can potentially be used for the treatment of breast cancer. This review highlights the advantages, mechanisms of action, stimuli-responsiveness properties, and recent advancements of hydrogels for treating breast cancer immunotherapy. Moreover, post-treatment and its clinical translations are discussed in this review. The integration of hydrogels in immunotherapy strategies may pave the way for more effective, personalized, and patient-friendly approaches to combat breast cancer, ultimately contributing to a brighter future for breast cancer patients.
Collapse
Affiliation(s)
- Jagannath Mondal
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea; Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea; Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - Kushal Chakraborty
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Edwin J Bunggulawa
- Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Vishnu Revuri
- Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, TX 79902, United States; Biomedical Engineering Program, College of Engineering, University of Texas at El Paso, El Paso, TX 79968, United States.
| | - Yong-Kyu Lee
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea; Department of Green Bioengineering, Korea National University of Transportation, Chungju 27470, Republic of Korea; Department of Chemical & Biological Engineering, Korea National University of Transportation, Chungju 27470, Republic of Korea.
| |
Collapse
|
20
|
Fan WL, Huang SY, Yang XJ, Bintang Ilhami F, Chen JK, Cheng CC. Hydrogen-bonded cytosine-endowed supramolecular polymeric nanogels: Highly efficient cancer cell targeting and enhanced therapeutic efficacy. J Colloid Interface Sci 2024; 665:329-344. [PMID: 38531278 DOI: 10.1016/j.jcis.2024.03.154] [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: 01/21/2024] [Revised: 03/10/2024] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
We demonstrate that cytosine moieties within physically cross-linked supramolecular polymers not only manipulate drug delivery and release, but also confer specific targeting of cancer cells to effectively enhance the safety and efficacy of chemotherapy-and thus hold significant potential as a new perspective for development of drug delivery systems. Herein, we successfully developed physically cross-linked supramolecular polymers (PECH-PEG-Cy) comprised of hydrogen-bonding cytosine pendant groups, hydrophilic poly(ethylene glycol) side chains, and a hydrophobic poly(epichlorohydrin) main chain. The polymers spontaneously self-assemble into a reversibly hydrogen-bonded network structure induced by cytosine and directly form spherical nanogels in aqueous solution. Nanogels with a high hydrogen-bond network density (i.e., a higher content of cytosine moieties) exhibit outstanding long-term structural stability in cell culture substrates containing serum, whereas nanogels with a relatively low hydrogen-bond network density cannot preserve their structural integrity. The nanogels also exhibit numerous unique physicochemical characteristics in aqueous solution, such as a desirable spherical size, high biocompatibility with normal and cancer cells, excellent drug encapsulation capacity, and controlled pH-responsive drug release properties. More importantly, in vitro experiments conclusively indicate the drug-loaded PECH-PEG-Cy nanogels can selectively induce cancer cell-specific apoptosis and cell death via cytosine receptor-mediated endocytosis, without significantly harming normal cells. In contrast, control drug-loaded PECH-PEG nanogels, which lack cytosine moieties in their structure, can only induce cell death in cancer cells through non-specific pathways, which significantly inhibits the induction of apoptosis. This work clearly demonstrates that the cytosine moieties in PECH-PEG-Cy nanogels confer selective affinity for the surface of cancer cells, which enhances their targeted cellular uptake, cytotoxicity, and subsequent induction of programmed cell death in cancer cells.
Collapse
Affiliation(s)
- Wen-Lu Fan
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Shan-You Huang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Xiu-Jing Yang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Fasih Bintang Ilhami
- Department of Natural Science, Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya, Surabaya 60231, Indonesia
| | - Jem-Kun Chen
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan
| | - Chih-Chia Cheng
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei 10607, Taiwan; Advanced Membrane Materials Research Center, National Taiwan University of Science and Technology, Taipei 10607, Taiwan.
| |
Collapse
|
21
|
Singhal R, Sarangi MK, Rath G. Injectable Hydrogels: A Paradigm Tailored with Design, Characterization, and Multifaceted Approaches. Macromol Biosci 2024; 24:e2400049. [PMID: 38577905 DOI: 10.1002/mabi.202400049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/22/2024] [Indexed: 04/06/2024]
Abstract
Biomaterials denoting self-healing and versatile structural integrity are highly curious in the biomedicine segment. The injectable and/or printable 3D printing technology is explored in a few decades back, which can alter their dimensions temporarily under shear stress, showing potential healing/recovery tendency with patient-specific intervention toward the development of personalized medicine. Thus, self-healing injectable hydrogels (IHs) are stunning toward developing a paradigm for tissue regeneration. This review comprises the designing of IHs, rheological characterization and stability, several benchmark consequences for self-healing IHs, their translation into tissue regeneration of specific types, applications of IHs in biomedical such as anticancer and immunomodulation, wound healing and tissue/bone regeneration, antimicrobial potentials, drugs, gene and vaccine delivery, ocular delivery, 3D printing, cosmeceuticals, and photothermal therapy as well as in other allied avenues like agriculture, aerospace, electronic/electrical industries, coating approaches, patents associated with therapeutic/nontherapeutic avenues, and numerous futuristic challenges and solutions.
Collapse
Affiliation(s)
- Rishika Singhal
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Manoj Kumar Sarangi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, 751030, India
| |
Collapse
|
22
|
Unnikrishnan Meenakshi D, Narde GK, Ahuja A, Al Balushi K, Francis AP, Khan SA. Therapeutic Applications of Nanoformulated Resveratrol and Quercetin Phytochemicals in Colorectal Cancer-An Updated Review. Pharmaceutics 2024; 16:761. [PMID: 38931884 PMCID: PMC11206904 DOI: 10.3390/pharmaceutics16060761] [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: 04/05/2024] [Revised: 05/07/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Natural compounds such as polyphenols play several positive roles in maintaining the oxidative and inflammatory capacity of cells, which leads to their potential use as anticancer therapeutics. There is promising evidence for the in vitro and in vivo anticancer activity of many polyphenols, including resveratrol and quercetin, specifically in the treatment of colorectal cancer (CRC). There is a clear association between resveratrol and quercetin in interfering with the mechanistic pathways involved in CRC, such as Wnt, P13K/AKT, caspase-3, MAPK, NF-κB, etc. These molecular pathways establish the role of resveratrol and quercetin in controlling cancer cell growth, inducing apoptosis, and inhibiting metastasis. The major bottleneck in the progression of the use of resveratrol and quercetin as anticancer therapeutics is their reduced bioavailability in vivo because of their rapid metabolism in humans. Recent advancements in various nanotechnological formulations are promising for overcoming these bioavailability issues. Various nanoformulations of resveratrol and quercetin have shown an optimistic impact on reducing the solubility and improving the stability of resveratrol and quercetin in vivo. A combinatorial approach using nanoformulations of resveratrol with quercetin could potentially increase the impact of resveratrol in controlling CRC cell proliferation. This review discusses the mechanism of resveratrol and quercetin, the two bioactive polyphenolics, in colon cancer, with an emphasis on various types of nanoformulations of the two molecules targeting colon cancer. It also explores the synergistic effect of combining resveratrol and quercetin in various nanoformulations, targeting colon cancer. This research delves into the enhanced pharmacokinetics and potential chemotherapeutic benefits of these bioactive polyphenolics when used together in innovative ways.
Collapse
Affiliation(s)
| | - Gurpreet Kaur Narde
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| | - Alka Ahuja
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| | - Khalid Al Balushi
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| | - Arul Prakash Francis
- Centre of Molecular Medicine and Diagnostics (COMMAND), Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India;
| | - Shah Alam Khan
- College of Pharmacy, National University of Science and Technology, Muscat PC 130, Oman; (G.K.N.); (K.A.B.); (S.A.K.)
| |
Collapse
|
23
|
Chen Y, Shu X, Guo JY, Xiang Y, Liang SY, Lai JM, Zhou JY, Liu LH, Wang P. Nanodrugs mediate TAMs-related arginine metabolism interference to boost photodynamic immunotherapy. J Control Release 2024; 367:248-264. [PMID: 38272398 DOI: 10.1016/j.jconrel.2024.01.045] [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: 08/25/2023] [Revised: 12/31/2023] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
As a potential treatment strategy for low immunogenic triple negative breast cancer (TNBC), photodynamic therapy (PDT) induced antitumor immunotherapy is greatly limited by the immunosuppressive tumor microenvironment (ITM), especially the M2 phenotype tumor-associated macrophages (TAMs). The balance of arginine metabolism plays an important role in TAMs polarization. Herein, a multifunctional nanoplatform (defined as HN-HFPA) was employed to burst the anti-tumor immunity of TNBC post PDT by reeducating TAMs through interfering the TAMs-associated arginine metabolism. The L-arginine (L-Arg) was loaded in the hollow cavity of HN-HFPA, which could not only generate nitric oxide (NO) for tumor therapy, but also serve as a substrate of arginine metabolism pathway. As an inhibitor of arginases-1 (Arg-1) of M2 TAMs, L-norvaline (L-Nor) was modified to the hyaluronic acid (HA), and coated in the surface of HFPA. After degradation of HA by hyaluronidase in tumor tissue and GSH-mediated disintegration, HN-HFPA depleted intracellular GSH, produced remarkable reactive oxygen species (ROS) under light irradiation and released L-Arg to generate NO, which induced tumor immunogenic cell death (ICD). Real-time ultrasound imaging of tumor was realized taking advantage of the gas feature of NO. The L-Nor suppressed the Arg-1 overexpressed in M2, which skewed the balance of arginine metabolism and reversed the ITM with increased ratios of M1 and CD8+ T cells, finally resulted in amplified antitumor immune response and apparent tumor metastasis inhibition. This study remodeled ITM to strengthen immune response post PDT, which provided a promising treatment strategy for TNBC.
Collapse
Affiliation(s)
- Yi Chen
- Department of Ultrasonography, The Third Affiliated Hospital, The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong Province, PR China
| | - Xian Shu
- Department of Ultrasonography, The Third Affiliated Hospital, The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong Province, PR China
| | - Jia-Yi Guo
- Department of Ultrasonography, The Third Affiliated Hospital, The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong Province, PR China
| | - Yun Xiang
- Department of Ultrasonography, The Third Affiliated Hospital, The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong Province, PR China
| | - Shi-Yu Liang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jin-Mei Lai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China
| | - Jia-Yi Zhou
- Department of Ultrasonography, The Third Affiliated Hospital, The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong Province, PR China
| | - Li-Han Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, PR China.
| | - Ping Wang
- Department of Ultrasonography, The Third Affiliated Hospital, The Third Clinical College, Southern Medical University, Guangzhou 510630, Guangdong Province, PR China.
| |
Collapse
|
24
|
Zhang H, Yuan W. Self-healable oxide sodium alginate/carboxymethyl chitosan nanocomposite hydrogel loading Cu 2+-doped MOF for enhanced synergistic and precise cancer therapy. Int J Biol Macromol 2024; 262:129996. [PMID: 38342271 DOI: 10.1016/j.ijbiomac.2024.129996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/25/2024] [Accepted: 02/04/2024] [Indexed: 02/13/2024]
Abstract
The limitations of traditional therapeutic methods such as chemotherapy serious restricted the application in tumor treatment, including poor targeting, toxic side effects and poor precision. It is important to develop non-chemotherapeutic systems to achieve precise and efficient tumor treatment. Therefore, a functional metal-organic framework material (MOF) with porphyrin core and doped with Cu2+ and surface-modified with polydopamine (PDA), namely PCN-224(Cu)@PDA (PCP) was designed and prepared. After loaded into the injectable and self-healable hydrogels by dynamic Schiff base bonding of oxidized sodium alginate (OSA) and carboxymethyl chitosan (CMC), the multifunctional nanocomposite hydrogels were obtained, in which Cu2+ in MOF converts to Cu+ by reacting with glutathione (GSH) which reduces the tumor antioxidant activity to improve the CDT effect. The Cu2+/Cu+ induces Fenton-like reaction in tumor cells to produce a toxic hydroxyl radical (OH). PDA achieves photothermal conversion under NIR light for photothermal therapy (PTT), and porphyrin core as a ligand generates reactive oxygen species (ROS), presenting highly efficient photodynamic therapy (PDT). Injectable self-healing hydrogel as a loading platform can be in situ injected to tumor site to release PCP and endocytosed by tumor cells to achieve precise and synergistic CDT-PDT-PTT therapy.
Collapse
Affiliation(s)
- Hanyan Zhang
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Weizhong Yuan
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China.
| |
Collapse
|
25
|
Shi H, Ma D, Wu D, Qiu X, Yang S, Wang Y, Xiao L, Ji X, Zhang W, Han S, Huo P, Dong J, Kong X, Guan X, Zhang D. A pH-responsive, injectable and self-healing chitosan-coumarin hydrogel based on Schiff base and hydrogen bonds. Int J Biol Macromol 2024; 255:128122. [PMID: 37984570 DOI: 10.1016/j.ijbiomac.2023.128122] [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: 05/07/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023]
Abstract
Smart hydrogels have shown great potential applications in disease treatment due to their controlled and local drug-release ability. Herein, a smart hydrogel with pH-responsive, injectable, and self-healing properties for controlled release of taxifolin (TFL) was prepared by freezing-thawing and photo-crosslinking methods. The crosslinking network of hydrogels (CS-CA hydrogels) was constructed by the hydrogen bonds, Schiff base bonds, and cyclobutane rings using chitosan (CS) and coumarin (CA) as raw materials. The CS-CA hydrogel demonstrated a compressive strength of 1.04 MPa, a self-healing efficiency of 99.9 %, and could maintain structural and functional integrity after injection. In addition, the drug release rate and shape of the CS-CA hydrogels were tunable due to its pH sensitivity. The TFL cumulative release reached 60 % within 12 h at pH = 4, and after equilibration, the cumulative release of TFL at pH = 4 (80 %) was significantly higher than at pH = 9.2 (50 %). The CCK8 experiment showed that the resulting hydrogel had no cytotoxicity. Meanwhile, subcutaneous implantation experiments in mice showed that the CS-CA hydrogels had favorable biodegradability and compatibility.
Collapse
Affiliation(s)
- Haolei Shi
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Dongxu Ma
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Di Wu
- Hospital of Northeast Forestry University, Northeast Forestry University, Harbin 150040, PR China
| | - Xiao Qiu
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Shuai Yang
- School of Materials Science and Engineering, North University of China, Taiyuan 030051, PR China
| | - Yingying Wang
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Lei Xiao
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Xinyao Ji
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Wei Zhang
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Shuaiyuan Han
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Pengfei Huo
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China
| | - Jidong Dong
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China.
| | - Xianzhi Kong
- Institute of Petrochemistry, Heilongjiang Academy of Sciences, Harbin 150040, PR China.
| | - Xue Guan
- Animal Laboratory Center, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, PR China.
| | - Dawei Zhang
- Engineering Research Center of Advanced Wooden Materials, Ministry of Education, Northeast Forestry University, Harbin 150040, PR China.
| |
Collapse
|
26
|
Cardador CM, de Castro TB, de Castro RJA, Bocca AL, Camargo LC, Pacheco TA, Muehlmann LA, Longo JPF. Doxorubicin-induced Immunogenic Cell Death Impairs Tumor Progression and Distant Metastasis in a 4T1 Breast Cancer Tumor Model. Curr Pharm Des 2024; 30:2493-2504. [PMID: 39041268 DOI: 10.2174/0113816128316870240610045550] [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: 03/18/2024] [Accepted: 05/09/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Cancer is an individual disease and its formation and development are specific to each host. Conventional treatments are ineffective in complex cases, such as metastasis, and have severe adverse side effects. New strategies are needed to address the problem, and the use of immunogenic cell death (ICD) as a trigger or booster of the immune system through the exposure of damage-associated molecular patterns, along with tumor antigens, by cancerous cells is presented as an immunization approach in this work. METHODS For this purpose, 4T1 cells were exposed to doxorubicin (DOX) for 24 hours and then, these cells undergoing ICD were subcutaneously administered to mice. The ICD induction by DOX on 4T1 was assessed by flow cytometry and image analysis. This immunization process was performed three times and after the last administration, the immunized mice were challenged with a subcutaneous xenograft of live cancer cells. RESULTS The results demonstrate that the mice immunized with cells undergoing ICD after exposure to DOX presented no primary tumor or indications of distant metastatic lesion development. CONCLUSION In summary, our findings indicate that the immunization process utilizing ICD is indeed efficacious in managing this aggressive form of pre-clinical breast cancer.
Collapse
Affiliation(s)
- Camila Magalhães Cardador
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Thaís Bergmann de Castro
- Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | | | - Anamélia Lorenzetti Bocca
- Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasília, Brazil
| | - Luana Cristina Camargo
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Thyago Arruda Pacheco
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| | - Luís Alexandre Muehlmann
- Department of Genetics and Morphology, Faculty of Ceilândia, University of Brasília, Brasília, Brazil
| | - João Paulo Figueiró Longo
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, Brazil
| |
Collapse
|
27
|
Zhang Z, He C, Chen X. Designing Hydrogels for Immunomodulation in Cancer Therapy and Regenerative Medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308894. [PMID: 37909463 DOI: 10.1002/adma.202308894] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/26/2023] [Indexed: 11/03/2023]
Abstract
The immune system not only acts as a defense against pathogen and cancer cells, but also plays an important role in homeostasis and tissue regeneration. Targeting immune systems is a promising strategy for efficient cancer treatment and regenerative medicine. Current systemic immunomodulation therapies are usually associated with low persistence time, poor targeting to action sites, and severe side effects. Due to their extracellular matrix-mimetic nature, tunable properties and diverse bioactivities, hydrogels are intriguing platforms to locally deliver immunomodulatory agents and cells, as well as provide an immunomodulatory microenvironment to recruit, activate, and expand host immune cells. In this review, the design considerations, including polymer backbones, crosslinking mechanisms, physicochemical nature, and immunomodulation-related components, of the hydrogel platforms, are focused on. The immunomodulatory effects and therapeutic outcomes in cancer therapy and tissue regeneration of different hydrogel systems are emphasized, including hydrogel depots for delivery of immunomodulatory agents, hydrogel scaffolds for cell delivery, and immunomodulatory hydrogels depending on the intrinsic properties of materials. Finally, the remained challenges in current systems and future development of immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Zhen Zhang
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, China
| |
Collapse
|
28
|
Singh S, Patil VM, Paliwal SK, Masand N. Nanotechnology-based Drug Delivery of Topical Antifungal Agents. Pharm Nanotechnol 2024; 12:185-196. [PMID: 37594096 DOI: 10.2174/2211738511666230818125031] [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: 05/16/2023] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Among the various prominent fungal infections, superficial ones are widespread. A large number of antifungal agents and their formulations for topical use are commercially available. They have some pharmacokinetic limitations which cannot be retracted by conventional delivery systems. While nanoformulations composed of lipidic and polymeric nanoparticles have the potential to overcome the limitations of conventional systems. The broad spectrum category of antifungals i.e. azoles (ketoconazole, voriconazole, econazole, miconazole, etc.) nanoparticles have been designed, prepared and their pharmacokinetic and pharmacodynamic profile was established. This review briefly elaborates on the types of nano-based topical drug delivery systems and portrays their advantages for researchers in the related field to benefit the available antifungal therapeutics.
Collapse
Affiliation(s)
- Sumita Singh
- Department of Pharmacy, Banasthali Vidyapith, Tonk, Rajasthan, India
- Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Vaishali M Patil
- Charak School of Pharmacy, Chaudhary Charan Singh (CCS) University, Meerut, Uttar Pradesh, India
| | | | - Neeraj Masand
- Department of Pharmacy, Lala Lajpat Rai Memorial Medical College, Meerut, Uttar Pradesh, India
| |
Collapse
|
29
|
Zhang T, Li N, Wang R, Sun Y, He X, Lu X, Chu L, Sun K. Enhanced therapeutic efficacy of doxorubicin against multidrug-resistant breast cancer with reduced cardiotoxicity. Drug Deliv 2023; 30:2189118. [PMID: 36919676 PMCID: PMC10026743 DOI: 10.1080/10717544.2023.2189118] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX), a commonly used anti-cancer drug, is limited by its cardiotoxicity and multidrug resistance (MDR) of tumor cells. Epigallocatechin gallate (EGCG), a natural antioxidant component, can effectively reduce the cardiotoxicity of DOX. Meanwhile, EGCG can inhibit the expression of P-glycoprotein (P-gp) and reverse the MDR of tumor cells. In this study, DOX is connected with low molecular weight polyethyleneimine (PEI) via hydrazone bond to get the pH-sensitive PEI-DOX, which is then combined with EGCG to prevent the cardiotoxicity of DOX and reverse the MDR of cancer cells. In addition, folic acid (FA) modified polyethylene glycol (PEG) (PEG-FA) is added to get the targeted system PEI-DOX/EGCG/FA. The MDR reversal and targeting ability of PEI-DOX/EGCG/FA is performed by cytotoxicity and in vivo anti-tumor activity on multidrug resistant MCF-7 cells (MCF-7/ADR). Additionally, we investigate the anti-drug resistant mechanism by Western Blot. The ability of EGCG to reduce DOX cardiotoxicity is confirmed by cardiotoxicity assay. In conclusion, PEI-DOX/EGCG/FA can inhibit the expression of P-gp and reverse the MDR in tumor cells. It also shows the ability of remove oxygen free radicals effectively to prevent the cardiotoxicity of DOX.
Collapse
Affiliation(s)
- Tianyu Zhang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Nuannuan Li
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Ru Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Yiying Sun
- Yantai Saipute Analyzing Service Co. Ltd, Yantai, Shandong Province, China
| | - Xiaoyan He
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Xiaoyan Lu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Liuxiang Chu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| | - Kaoxiang Sun
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Ministry of Education, Yantai University, Yantai, China
| |
Collapse
|
30
|
Mohaghegh N, Ahari A, Zehtabi F, Buttles C, Davani S, Hoang H, Tseng K, Zamanian B, Khosravi S, Daniali A, Kouchehbaghi NH, Thomas I, Serati Nouri H, Khorsandi D, Abbasgholizadeh R, Akbari M, Patil R, Kang H, Jucaud V, Khademhosseini A, Hassani Najafabadi A. Injectable hydrogels for personalized cancer immunotherapies. Acta Biomater 2023; 172:67-91. [PMID: 37806376 DOI: 10.1016/j.actbio.2023.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/19/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
The field of cancer immunotherapy has shown significant growth, and researchers are now focusing on effective strategies to enhance and prolong local immunomodulation. Injectable hydrogels (IHs) have emerged as versatile platforms for encapsulating and controlling the release of small molecules and cells, drawing significant attention for their potential to enhance antitumor immune responses while inhibiting metastasis and recurrence. IHs delivering natural killer (NK) cells, T cells, and antigen-presenting cells (APCs) offer a viable method for treating cancer. Indeed, it can bypass the extracellular matrix and gradually release small molecules or cells into the tumor microenvironment, thereby boosting immune responses against cancer cells. This review provides an overview of the recent advancements in cancer immunotherapy using IHs for delivering NK cells, T cells, APCs, chemoimmunotherapy, radio-immunotherapy, and photothermal-immunotherapy. First, we introduce IHs as a delivery matrix, then summarize their applications for the local delivery of small molecules and immune cells to elicit robust anticancer immune responses. Additionally, we discuss recent progress in IHs systems used for local combination therapy, including chemoimmunotherapy, radio-immunotherapy, photothermal-immunotherapy, photodynamic-immunotherapy, and gene-immunotherapy. By comprehensively examining the utilization of IHs in cancer immunotherapy, this review aims to highlight the potential of IHs as effective carriers for immunotherapy delivery, facilitating the development of innovative strategies for cancer treatment. In addition, we demonstrate that using hydrogel-based platforms for the targeted delivery of immune cells, such as NK cells, T cells, and dendritic cells (DCs), has remarkable potential in cancer therapy. These innovative approaches have yielded substantial reductions in tumor growth, showcasing the ability of hydrogels to enhance the efficacy of immune-based treatments. STATEMENT OF SIGNIFICANCE: As cancer immunotherapy continues to expand, the mode of therapeutic agent delivery becomes increasingly critical. This review spotlights the forward-looking progress of IHs, emphasizing their potential to revolutionize localized immunotherapy delivery. By efficiently encapsulating and controlling the release of essential immune components such as T cells, NK cells, APCs, and various therapeutic agents, IHs offer a pioneering pathway to amplify immune reactions, moderate metastasis, and reduce recurrence. Their adaptability further shines when considering their role in emerging combination therapies, including chemoimmunotherapy, radio-immunotherapy, and photothermal-immunotherapy. Understanding IHs' significance in cancer therapy is essential, suggesting a shift in cancer treatment dynamics and heralding a novel period of focused, enduring, and powerful therapeutic strategies.
Collapse
Affiliation(s)
- Neda Mohaghegh
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Amir Ahari
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Claire Buttles
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Indiana University Bloomington, Department of Biology, Bloomington, IN 47405, USA
| | - Saya Davani
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Hanna Hoang
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA 90024, USA
| | - Kaylee Tseng
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90007, USA
| | - Benjamin Zamanian
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Electrical and Computer Engineering, University of British Columbia, Vancouver, BC, V6T1Z4, Canada
| | - Ariella Daniali
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Negar Hosseinzadeh Kouchehbaghi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Department of Textile Engineering, Amirkabir University of Technology (Tehran Polytechnic), Hafez Avenue, Tehran, Iran
| | - Isabel Thomas
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA
| | - Hamed Serati Nouri
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohsen Akbari
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA; Laboratory for Innovations in Microengineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Rameshwar Patil
- Department of Basic Science and Neurosurgery, Division of Cancer Science, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Heemin Kang
- Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064 USA.
| | | |
Collapse
|
31
|
Gao Y, Zhang Y, Xia H, Ren Y, Zhang H, Huang S, Li M, Wang Y, Li H, Liu H. Biomimetic virus-like mesoporous silica nanoparticles improved cellular internalization for co-delivery of antigen and agonist to enhance Tumor immunotherapy. Drug Deliv 2023; 30:2183814. [PMID: 36843529 PMCID: PMC9980018 DOI: 10.1080/10717544.2023.2183814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2023] Open
Abstract
Nanocarrier antigen-drug delivery system interacts specifically with immune cells and provides intelligent delivery modes to improve antigen delivery efficiency and facilitate immune progression. However, these nanoparticles often have weak adhesion to cells, followed by insufficient cell absorption, leading to a failed immune response. Inspired by the structure and function of viruses, virus-like mesoporous silica nanoparticles (VMSNs) were prepared by simulating the surface structure, centripetal-radialized spike structure and rough surface topology of the virus and co-acted with the toll-like receptor 7/8 agonist imiquimod (IMQ) and antigens oocyte albumin (OVA). Compared to the conventional spherical mesoporous silica nanoparticles (MSNs), VMSNs which was proven to be biocompatible in both cellular and in vivo level, had higher cell invasion ability and unique endocytosis pathway that was released from lysosomes and promoted antigen cross-expression. Furthermore, VMSNs effectively inhibited B16-OVA tumor growth by activating DCs maturation and increasing the proportion of CD8+ T cells. This work demonstrated that virus-like mesoporous silica nanoparticles co-supply OVA and IMQ, could induce potent tumor immune responses and inhibit tumor growth as a consequence of the surface spike structure induces a robust cellular immune response, and undoubtedly provided a good basis for further optimizing the nanovaccine delivery system.
Collapse
Affiliation(s)
- Yuan Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yingxi Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Hong Xia
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuqing Ren
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Haibin Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Siwen Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Meiju Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Heran Li
- School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Hongzhuo Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China,CONTACT Heran Li School of Pharmacy, China Medical University, 77 Puhe Road, Shenyang North New Area, Liaoning, 110122, China; Hongzhuo Liu Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| |
Collapse
|
32
|
Stepanova M, Nikiforov A, Tennikova T, Korzhikova-Vlakh E. Polypeptide-Based Systems: From Synthesis to Application in Drug Delivery. Pharmaceutics 2023; 15:2641. [PMID: 38004619 PMCID: PMC10674432 DOI: 10.3390/pharmaceutics15112641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/02/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Synthetic polypeptides are biocompatible and biodegradable macromolecules whose composition and architecture can vary over a wide range. Their unique ability to form secondary structures, as well as different pathways of modification and biofunctionalization due to the diversity of amino acids, provide variation in the physicochemical and biological properties of polypeptide-containing materials. In this review article, we summarize the advances in the synthesis of polypeptides and their copolymers and the application of these systems for drug delivery in the form of (nano)particles or hydrogels. The issues, such as the diversity of polypeptide-containing (nano)particle types, the methods for their preparation and drug loading, as well as the influence of physicochemical characteristics on stability, degradability, cellular uptake, cytotoxicity, hemolysis, and immunogenicity of polypeptide-containing nanoparticles and their drug formulations, are comprehensively discussed. Finally, recent advances in the development of certain drug nanoformulations for peptides, proteins, gene delivery, cancer therapy, and antimicrobial and anti-inflammatory systems are summarized.
Collapse
Affiliation(s)
- Mariia Stepanova
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy pr. 31, 199004 St. Petersburg, Russia; (M.S.); (A.N.)
| | - Alexey Nikiforov
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy pr. 31, 199004 St. Petersburg, Russia; (M.S.); (A.N.)
| | - Tatiana Tennikova
- Institute of Chemistry, Saint-Petersburg State University, Universitetskiy pr. 26, Petergof, 198504 St. Petersburg, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Macromolecular Compounds, Russian Academy of Sciences, Bolshoy pr. 31, 199004 St. Petersburg, Russia; (M.S.); (A.N.)
| |
Collapse
|
33
|
Pelin IM, Popescu I, Calin M, Rebleanu D, Voicu G, Ionita D, Zaharia MM, Constantin M, Fundueanu G. Tri-Component Hydrogel as Template for Nanocrystalline Hydroxyapatite Deposition Using Alternate Soaking Method for Bone Tissue Engineering Applications. Gels 2023; 9:905. [PMID: 37998995 PMCID: PMC10671408 DOI: 10.3390/gels9110905] [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: 09/27/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
Composite hydrogels containing apatite-like particles can act as scaffolds for osteoblast proliferation, with applications in bone tissue engineering. In this respect, porous biocompatible hydrogels were obtained from chitosan, oxidized pullulan, and PVA in different ratios. The stability of the hydrogels was ensured both by covalent bonds between aldehyde groups of oxidized pullulan and free amino groups of chitosan, and by physical bonds formed during freeze-thaw cycles and lyophilization. The deposition of calcium phosphates was performed by alternate soaking of the porous hydrogels into solutions with calcium and phosphate ions, assuring a basic pH required for hydroxyapatite formation. The mineralized hydrogels were characterized using FTIR spectroscopy, scanning electron microscopy, X-ray diffraction, and thermogravimetric analysis, showing that inorganic particles containing between 80 and 92% hydroxyapatite were deposited in a high amount on the pore walls of the polymeric matrix. The composition of the organic matrix influenced the crystallization of calcium phosphates and the mechanical properties of the composite hydrogels. In vitro biological tests showed that mineralized hydrogels support the proliferation of MG-63 osteoblast-like cells to a greater extent compared to pristine hydrogels.
Collapse
Affiliation(s)
- Irina Mihaela Pelin
- “Petru Poni” Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley 41A, 700487 Iasi, Romania; (I.M.P.); (D.I.); (M.-M.Z.); (G.F.)
| | - Irina Popescu
- “Petru Poni” Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley 41A, 700487 Iasi, Romania; (I.M.P.); (D.I.); (M.-M.Z.); (G.F.)
| | - Manuela Calin
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.C.); (D.R.); (G.V.)
| | - Daniela Rebleanu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.C.); (D.R.); (G.V.)
| | - Geanina Voicu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania; (M.C.); (D.R.); (G.V.)
| | - Daniela Ionita
- “Petru Poni” Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley 41A, 700487 Iasi, Romania; (I.M.P.); (D.I.); (M.-M.Z.); (G.F.)
| | - Marius-Mihai Zaharia
- “Petru Poni” Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley 41A, 700487 Iasi, Romania; (I.M.P.); (D.I.); (M.-M.Z.); (G.F.)
| | - Marieta Constantin
- “Petru Poni” Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley 41A, 700487 Iasi, Romania; (I.M.P.); (D.I.); (M.-M.Z.); (G.F.)
| | - Gheorghe Fundueanu
- “Petru Poni” Institute of Macromolecular Chemistry, Grigore Ghica Voda Alley 41A, 700487 Iasi, Romania; (I.M.P.); (D.I.); (M.-M.Z.); (G.F.)
| |
Collapse
|
34
|
Cao Y, Sun T, Sun B, Zhang G, Liu J, Liang B, Zheng C, Kan X. Injectable hydrogel loaded with lysed OK-432 and doxorubicin for residual liver cancer after incomplete radiofrequency ablation. J Nanobiotechnology 2023; 21:404. [PMID: 37919724 PMCID: PMC10623833 DOI: 10.1186/s12951-023-02170-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 10/18/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE To investigate the efficacy of an injectable hydrogel loaded with lysed OK-432 (lyOK-432) and doxorubicin (DOX) for residual liver cancer after incomplete radiofrequency ablation (iRFA) of hepatocellular carcinoma (HCC), and explore the underlying mechanism. MATERIALS AND METHODS The effect of OK-432 and lyOK-432 was compared in activating dendritic cells (DCs). RADA16-I (R) peptide was dissolved in a mixture of lyOK-432 (O) and DOX (D) to develop an ROD hydrogel. The characteristics of ROD hydrogel were evaluated. Tumor response and mice survival were measured after different treatments. The number of immune cells and cytokine levels were measured, and the activation of cGAS/STING/IFN-I signaling pathway in DC was evaluated both in vitro and in vivo. RESULTS LyOK-432 was more effective than OK-432 in promoting DC maturation and activating the IFN-I pathway. ROD was an injectable hydrogel for effectively loading lyOK-432 and DOX, and presented the controlled-release property. ROD treatment achieved the highest tumor necrosis rate (p < 0.001) and the longest survival time (p < 0.001) compared with the other therapies. The ROD group also displayed the highest percentages of DCs, CD4+ T cells and CD8+ T cells (p < 0.001), the lowest level of Treg cells (p < 0.001), and the highest expression levels of IFN-γ and TNF-α (p < 0.001) compared with the other groups. The expression levels of pSTING, pIRF3, and IFN-β in DCs were obviously higher after treatment of lyOK-432 in combination with DOX than the other therapies. The surviving mice in the ROD group showed a growth inhibition of rechallenged subcutaneous tumor. CONCLUSION The novel ROD peptide hydrogel induced an antitumor immunity by activating the STING pathway, which was effective for treating residual liver cancer after iRFA of HCC.
Collapse
Affiliation(s)
- Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tao Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bo Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Guilin Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Jiayun Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China.
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China.
| |
Collapse
|
35
|
Yu K, Ye B, Yang H, Xu X, Mao Z, Zhang Q, Tian M, Zhang H, Zhang H, He Q. A Mitochondria-Targeted NIR-II AIEgen Induced Pyroptosis for Enhanced Tumor Immunotherapy. Adv Healthc Mater 2023; 12:e2301693. [PMID: 37285905 DOI: 10.1002/adhm.202301693] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Indexed: 06/09/2023]
Abstract
Cancer immunotherapy is a favorable strategy for facilitating anti-tumor immunity, but it shows limited benefits in clinical practice owing to the immunosuppressive tumor microenvironment. Pyroptosis shows great immunostimulatory effect on tumor, whereas the lack of pyroptotic inducer with imaging property has restricted its progress in tumor theranostics. Herein, a mitochondria-targeted aggregation-induced emission (AIE) luminogen (TPA-2TIN) with NIR-II emission is designed for highly efficient induction of tumor cell pyroptosis. The fabricated TPA-2TIN nanoparticles can be efficiently taken up by tumor cells and selectively accumulated in tumor for a long term observed by NIR-II fluorescence imaging. More importantly, the TPA-2TIN nanoparticles can effectively stimulate immune responses both in vitro and in vivo mediated by the mitochondrial dysfunctions and the subsequent activation of the pyroptotic pathway. Ultimately, the reversal of the immunosuppressive tumor microenvironment significantly enhances the immune checkpoint therapy. This study paves a new avenue for adjuvant immunotherapy of cancer.
Collapse
Affiliation(s)
- Kaiwu Yu
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Binglin Ye
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
| | - Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Xinxin Xu
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, P. R. China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Qinghua Zhang
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Mei Tian
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P. R. China
| | - Haoke Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Hong Zhang
- Department of Nuclear Medicine and PET Center, The Second Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, P. R. China
| | - Qinggang He
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
36
|
Perveen M, Noreen L, Waqas M, Mehmood RF, Iqbal J, Manzoor S, Nazir S, Shawky AM, Khera RA. A DFT approach for finding therapeutic potential of graphyne as a nanocarrier in the doxorubicin drug delivery to treat cancer. J Mol Graph Model 2023; 124:108537. [PMID: 37321062 DOI: 10.1016/j.jmgm.2023.108537] [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: 03/13/2023] [Revised: 05/19/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023]
Abstract
In the present work, the drug-loading efficacy of graphyne (GYN) for doxorubicin (DOX) drug is investigated for the first time by using density functional theory (DFT). Doxorubicin drug is effective in the cure of numerous types of cancer including bone cancer, gastric, thyroid, bladder, ovarian, breast, and soft tissue cancer. Doxorubicin drug prevents the cell division process by intercalating in the double-helix of DNA and stopping its replication. The optimized, geometrical, energetic, and excited-state characteristics of graphyne (GYN), doxorubicin drug (DOX), and doxorubicin-graphyne complex (DOX@GYN complex) are calculated to see how effective it is as a carrier. The DOX drug interacted with GYN with an adsorption-energy of -1.57 eV (gas-phase). The interaction of GYN with DOX drug is investigated using NCI (non-covalent interaction) analysis. The findings of this analysis showed that the DOX@GYN complex has weak forces of interaction. Charge transfer from doxorubicin drug to GYN during DOX@GYN complex formation is described by charge-decomposition analysis and HOMO-LUMO analysis. The increased dipole-moment (8.41 D) of the DOX@GYN in contrast with therapeutic agent DOX and GYN indicated that the drug will move easily in the biochemical system. Furthermore, the photo-induced electron-transfer process is explored for excited states, and it reveals that upon interaction, fluorescence-quenching will occur in the complex DOX@GYN. In addition, the influence of the positive and negative charge states on the GYN and DOX@GYN is also considered. Overall, the findings indicated that the GYN could be exploited as an effective drug-transporter for the delivery of doxorubicin drug. Investigators will be inspired to look at another 2D nanomaterials for drug transport applications as a result of this theoretical work.
Collapse
Affiliation(s)
- Mehvish Perveen
- Department of Chemistry, University of Agriculture, 38000 Faisalabad, Pakistan
| | - Lubna Noreen
- Department of Chemistry, University of Agriculture, 38000 Faisalabad, Pakistan
| | - Muhammad Waqas
- Department of Chemistry, University of Agriculture, 38000 Faisalabad, Pakistan
| | - Rana Farhat Mehmood
- Department of Chemistry, University of Education, Township, Lahore, 54770, Pakistan
| | - Javed Iqbal
- Department of Chemistry, University of Agriculture, 38000 Faisalabad, Pakistan.
| | - Sidra Manzoor
- Department of Chemistry, University of Agriculture, 38000 Faisalabad, Pakistan
| | - Sidra Nazir
- Faisalabad Institute of Cardiology, Faisalabad, Pakistan
| | - Ahmed M Shawky
- Science and Technology Unit (STU), Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Rasheed Ahmad Khera
- Department of Chemistry, University of Agriculture, 38000 Faisalabad, Pakistan.
| |
Collapse
|
37
|
Erdogan MA, Ugo D, Ines F. The role of ion channels in the relationship between the immune system and cancer. CURRENT TOPICS IN MEMBRANES 2023; 92:151-198. [PMID: 38007267 DOI: 10.1016/bs.ctm.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
The immune system is capable of identifying and eliminating cancer, a complicated illness marked by unchecked cellular proliferation. The significance of ion channels in the complex interaction between the immune system and cancer has been clarified by recent studies. Ion channels, which are proteins that control ion flow across cell membranes, have variety of physiological purposes, such as regulating immune cell activity and tumor development. Immune cell surfaces contain ion channels, which have been identified to control immune cell activation, motility, and effector activities. The regulation of immune responses against cancer cells has been linked to a number of ion channels, including potassium, calcium, and chloride channels. As an example, potassium channels are essential for regulating T cell activation and proliferation, which are vital for anti-tumor immunity. Calcium channels play a crucial role when immune cells produce cytotoxic chemicals in order to eliminate cancer cells. Chloride channels also affect immune cell infiltration and invasion into malignancies. Additionally, tumor cells' own expressed ion channels have an impact on their behavior and in the interaction with the immune system. The proliferation, resistance to apoptosis, and immune evasion of cancer cells may all be impacted by changes in ion channel expression and function. Ion channels may also affect the tumor microenvironment by controlling angiogenesis, inflammatory responses, and immune cell infiltration. Ion channel function in the interaction between the immune system and cancer has important implications for cancer treatment. A possible method to improve anti-tumor immune responses and stop tumor development is to target certain ion channels. Small compounds and antibodies are among the ion channel modulators under investigation as possible immunotherapeutics. The complex interaction between ion channels, the immune system, and cancer highlights the significance of these channels for tumor immunity. The development of novel therapeutic strategies for the treatment of cancer will be made possible by unraveling the processes by which ion channels control immune responses and tumor activity. Hence, the main driving idea of the present chapter is trying to understand the possible function of ion channels in the complex crosstalk between cancer and immunoresponse. To this aim, after giving a brief journey of ion channels throughout the history, a classification of the main ion channels involved in cancer disease will be discussed. Finally, the last paragraph will focus on more recently advancements in the use of biomaterials as therapeutic strategy for cancer treatment. The hope is that future research will take advantage of the promising combination of ion channels, immunomodulation and biomaterials filed to provide better solutions in the treatment of cancer disease.
Collapse
Affiliation(s)
- Mumin Alper Erdogan
- Izmir Katip Celebi University Faculty of Medicine, Department of Physiology, Izmir, Turkey.
| | - D'Amora Ugo
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, Italy
| | - Fasolino Ines
- Institute of Polymers, Composites and Biomaterials - National Research Council (IPCB-CNR), Naples, Italy
| |
Collapse
|
38
|
Choi W, Shin WR, Kim YH, Min J. Inducing a Proinflammatory Response with Bioengineered Yeast Vacuoles with TLR2-Binding Peptides (Vac T2BP) as a Drug Carrier for Daunorubicin Delivery. ACS APPLIED MATERIALS & INTERFACES 2023; 15:41258-41270. [PMID: 37615983 DOI: 10.1021/acsami.3c06669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Immune adjuvants have roles in immune activation for cancer therapy, and adjuvants derived from microbes have been applied. In this study, we propose the use of bioengineered vacuoles, derived from recombinant yeast with acute myeloid leukemia (AML) specificity and having a TLR-2-binding peptide (VacT2BP) on their surface, to induce a proinflammatory response as a dual-function nanomaterial for daunorubicin (DNR) delivery. Our results demonstrate that nanosized, isolated VacT2BP induced HL-60 cell-specific DNR delivery and apoptosis. Furthermore, we observed the selective release of high-mobility group box 1 from apoptotic HL-60 cells by DNR@VacT2BP. We concluded that DNR@VacT2BP exhibited target selectivity, and the indiscriminate occurrence of damage-associated molecular patterns (DAMPs) was inhibited by the VacT2BP carrier. The therapeutic efficacy of DNR@VacT2BP was confirmed in AML xenograft mice, with about 82% tumor growth inhibition. Following drug delivery, apoptotic cells and DAMPs with residual VacT2BP (apopDNR@VacT2BP) upregulated the proinflammatory immune response of macrophages. In addition, apopDNR@VacT2BP enhanced phagocytosis activity. Macrophages stimulated by apopDNR@VacT2BP suppressed cancer proliferation by about 40%. In summary, our results suggest that dual-functional vacuoles with a target-specific peptide can be a potential strategy for selective drug delivery and construction of an immune environment to fight cancer, thereby improving prognosis.
Collapse
Affiliation(s)
- Wooil Choi
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| | - Woo-Ri Shin
- School of Biological Sciences, Chungbuk National University, 1, Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Yang-Hoon Kim
- School of Biological Sciences, Chungbuk National University, 1, Chungdae-Ro, Seowon-Gu, Cheongju 28644, South Korea
| | - Jiho Min
- Graduate School of Semiconductor and Chemical Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-Gu Jeonju, Jeonbuk 54896, South Korea
| |
Collapse
|
39
|
Fang T, Cao X, Shen B, Chen Z, Chen G. Injectable cold atmospheric plasma-activated immunotherapeutic hydrogel for enhanced cancer treatment. Biomaterials 2023; 300:122189. [PMID: 37307777 DOI: 10.1016/j.biomaterials.2023.122189] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/19/2023] [Accepted: 05/31/2023] [Indexed: 06/14/2023]
Abstract
Despite the promise of immune checkpoint blockade (ICB) for cancer treatment, challenges associated with this therapy still exist, including low response rates and severe side effects in patients. Here, we report a hydrogel-mediated combination therapy for enhanced ICB therapy. Specifically, cold atmospheric plasma (CAP), an ionized gas consisting of therapeutically effective reactive oxygen species (ROS) and reactive nitrogen species (RNS), can effectively induce cancer immunogenic cell death, releasing tumor-associated antigens in situ and initiating anti-tumor immune responses, which, therefore, can synergistically augment the efficacy of immune checkpoint inhibitors. To minimize the systemic toxicity of immune checkpoint inhibitors and improve the tissue penetration of CAP, an injectable Pluronic hydrogel was employed as a delivery method. Our results show that major long-lived ROS and RNS in CAP can be effectively persevered in Pluronic hydrogel and remain efficacious in inducing cancer immunogenic cell death after intratumoral injection. Our findings suggest that local hydrogel-mediated combination of CAP and ICB treatment can evoke both strong innate and adaptive, local and systemic anti-tumor immune responses, thereby inhibiting both tumor growth and potential metastatic spread.
Collapse
Affiliation(s)
- Tianxu Fang
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Xiaona Cao
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada; School of Nursing, Tianjin Medical University, Tianjin, China
| | - Bingzheng Shen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Zhitong Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Center for Advanced Therapy, National Innovation Center for Advanced Medical Devices, Shenzhen, China.
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada.
| |
Collapse
|
40
|
Erfani A, Reichert P, Narasimhan CN, Doyle PS. Injectable hydrogel particles for amorphous solid formulation of biologics. iScience 2023; 26:107452. [PMID: 37593455 PMCID: PMC10428138 DOI: 10.1016/j.isci.2023.107452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/19/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023] Open
Abstract
The fast pace of breakthroughs in cancer immunotherapy, combined with the new paradigm of moving toward high-concentration dosages and combinatorial treatments, is generating new challenges in the formulation of biologics. To address these challenges, we describe a method of formulation that enables high-concentration injectable and stable formulation of biologics as amorphous solids in aqueous suspension. This technology combines the benefits of liquid formulation with the stability of solid formulation and eliminates the need for drying and reconstitution. This widely applicable formulation integrates the amorphous solid forms of antibodies with the injectability, lubricity, and tunability of soft alginate hydrogel particles using a minimal process. The platform was evaluated for anti-PD-1 antibody pembrolizumab and human immunoglobulin G at concentrations up to 300 mg/mL with confirmed quality after release. The soft nature of the hydrogel matrix allowed packing the particles to high volume fractions.
Collapse
Affiliation(s)
- Amir Erfani
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | | | - Patrick S. Doyle
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Harvard Medical School Initiative for RNA Medicine, Boston, MA 02215, USA
| |
Collapse
|
41
|
Ahmad I, Farheen M, Kukreti A, Afzal O, Akhter MH, Chitme H, Visht S, Altamimi AS, Alossaimi MA, Alsulami ER, Jaremko M, Emwas AH. Natural Oils Enhance the Topical Delivery of Ketoconazole by Nanoemulgel for Fungal Infections. ACS OMEGA 2023; 8:28233-28248. [PMID: 37576685 PMCID: PMC10413480 DOI: 10.1021/acsomega.3c01571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/08/2023] [Indexed: 08/15/2023]
Abstract
Nanoemulgel (NEG) pharmaceutical formulations are gaining popularity because of their ability to serve both as a nanoemulsion and as a gel. These products are well-known for their ease of use, spreadability, controlled release, and ability to hydrate dry skin. Natural essential oils have been shown to promote the cutaneous permeability of topical formulations, enhancing medication safety and efficacy. Herein, we developed NEG for the enhanced permeation of ketoconazole against candidiasis using clove oil (clove-oil-NEG) or eucalyptus oil (eucalyptus-oil-NEG), using the gelling agents carbopol 943 and hydroxypropyl methylcellulose (HPMC). We tested various excipients to increase the solubility of ketoconazole and formulate a nanoemulsion (NE). We measured the NE droplet particle size, shape, entrapment efficiency, and drug release. Furthermore, the physicochemical properties of the optimized nanoemulsion formulation were characterized by techniques such as Fourier transform infrared (FT-IR) spectroscopy and X-ray diffraction (XRD) analysis. The NEs were loaded into gels to form NEGs. NEGs were characterized for drug content, homogeneity, rheology, spreadability, and antifungal activity against Candida albicans, both in vitro and in vivo. Optimized ketoconazole NEG preparations consisted of either 15% clove oil or 20% eucalyptus oil. Droplet sizes in the optimized NEs were <100 nm, and the polydispersity indexes were 0.24 and 0.26. The percentages of ketoconazole released after 24 h from the clove-oil-NEG and eucalyptus-oil-NEGs were 91 ± 4.5 and 89 ± 7%, respectively. Scanning electron microscopy (SEM) showed that the NEGs had a smooth, uniform, and consistent shape and internal structural organization. The drug contents in the clove-oil-NEG and eucalyptus-oil-NEG were 98.5 ± 2.2 and 98.8 ± 3.4%, respectively. Permeation values of ketoconazole from clove-oil-NEG and eucalyptus-oil-NEG were 117 ± 7 and 108.34 ± 6 μg cm-2, respectively. The ketoconazole NEG formulations also had higher levels of fungal growth inhibition than a marketed formulation. Finally, in vivo studies showed that the NEGs do not irritate the skin. Ketoconazole NEG with either 15% clove oil or 20% eucalyptus oil is stable with better efficacy than ketoconazole alone due to excellent dispersion, drug dissolution, and permeability and thus might be recommended for the effective and safe treatment of candidiasis.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62521, Saudi Arabia
| | - Ms Farheen
- School
of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Ashish Kukreti
- School
of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Obaid Afzal
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Md Habban Akhter
- School
of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Havagiray Chitme
- School
of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Sharad Visht
- School
of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | | | - Manal A. Alossaimi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Ebtisam R. Alsulami
- Nursing
Department, Najran Armed Forces Hospital, Najran 66251, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health
Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological
and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology
(KAUST), Thuwal 23955, Saudi Arabia
| |
Collapse
|
42
|
Omidian H, Chowdhury SD. High-Performing Conductive Hydrogels for Wearable Applications. Gels 2023; 9:549. [PMID: 37504428 PMCID: PMC10379850 DOI: 10.3390/gels9070549] [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: 06/21/2023] [Revised: 07/04/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023] Open
Abstract
Conductive hydrogels have gained significant attention for their extensive applications in healthcare monitoring, wearable sensors, electronic devices, soft robotics, energy storage, and human-machine interfaces. To address the limitations of conductive hydrogels, researchers are focused on enhancing properties such as sensitivity, mechanical strength, electrical performance at low temperatures, stability, antibacterial properties, and conductivity. Composite materials, including nanoparticles, nanowires, polymers, and ionic liquids, are incorporated to improve the conductivity and mechanical strength. Biocompatibility and biosafety are emphasized for safe integration with biological tissues. Conductive hydrogels exhibit unique properties such as stretchability, self-healing, wet adhesion, anti-freezing, transparency, UV-shielding, and adjustable mechanical properties, making them suitable for specific applications. Researchers aim to develop multifunctional hydrogels with antibacterial characteristics, self-healing capabilities, transparency, UV-shielding, gas-sensing, and strain-sensitivity.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Sumana Dey Chowdhury
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
43
|
Zaboli A, Raissi H, Hashemzadeh H, Farzad F. Graphene Oxide Hosting a pH-Sensitive Prodrug: An In Silico Investigation of Graphene Oxide-Based Nanovehicle toward Cancer Therapy. ACS APPLIED BIO MATERIALS 2023. [PMID: 37327458 DOI: 10.1021/acsabm.3c00276] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Prodrug and drug delivery systems are two effective strategies for improving the selectivity of chemotherapeutics. Herein, via molecular dynamics (MD) simulation and free energy calculation, the effectiveness of the graphene oxide (GO) decorated with the pH-sensitive prodrug (PD) molecules in cancer therapy is investigated. PEI-CA-DOX (prodrug) was loaded onto the GO surface, in which the hydrogen bonding and pi-pi stacking interactions play the main role in the stability of the GO-PD complex. Due to the strong interaction of GO and PD (about -800 kJ/mol), the GO-PD complex remains stable during the membrane penetration process. The obtained results confirm that GO is a suitable surface for hosting the prodrug and passing it through the membrane. Furthermore, the investigation of the release process shows that the PD can be released under acidic conditions. This phenomenon is due to the reduction of the contribution of electrostatic energy in the GO and PD interaction and the entry of water into the drug delivery system. Moreover, it is found that an external electrical field does not have much effect on drug release. Our results provide a deep understanding of the prodrug delivery systems, which helps the combination of nanocarriers and modified chemotherapy drugs in the future.
Collapse
Affiliation(s)
- Ameneh Zaboli
- Department of Chemistry, University of Birjand, Birjand 9717434765, Iran
| | - Heidar Raissi
- Department of Chemistry, University of Birjand, Birjand 9717434765, Iran
| | - Hassan Hashemzadeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Birjand University of Medical Sciences, Birjand 9717853076, Iran
| | - Farzaneh Farzad
- Department of Chemistry, University of Birjand, Birjand 9717434765, Iran
| |
Collapse
|
44
|
Dang W, Xing B, Jia X, Zhang Y, Jia B, Yu C, He J, Li Z, Li H, Liu Z. Subcellular Organelle-Targeted Nanostructured Lipid Carriers for the Treatment of Metastatic Breast Cancer. Int J Nanomedicine 2023; 18:3047-3068. [PMID: 37312934 PMCID: PMC10259594 DOI: 10.2147/ijn.s413680] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/21/2023] [Indexed: 06/15/2023] Open
Abstract
Background Subcellular organelle targeted nano-formulations for cancer treatment are receiving increasing attention owing to their benefits of precise drug delivery, maximized therapeutic index, and reduced off-target side effects. The nucleus and mitochondria, as the main subcellular organelles, are the significant organelles responsible for maintaining cell operation and metabolism. They can be involved in many essential physiological and pathological processes such as cell proliferation, organism metabolism, intracellular transportation, and play a critical role in regulating cell biology. Meanwhile, breast cancer metastasis is one of the leading causes of death in breast cancer patients. With the development of nanotechnology, nanomaterials have been widely used in tumor therapy. Methods We designed a subcellular organelle targeted nanostructured lipid carriers (NLC) to deliver paclitaxel (PTX) and gambogic acid (GA) to tumor tissues. Results Due to the surface of NLC being modified by subcellular organelle targeted peptide, the PTX and GA co-loaded NLC can accurately release PTX and GA in tumor cells. This property makes NLC able to easy to enter tumor site and target the specific subcellular organelle. The modified NLC can efficiently inhibit the growth of 4T1 primary tumor and lung metastasis, which may be related to the down-regulation of matrix metalloproteinase-9 (MMP-9) and BCL-2 levels, up-regulation of E-cadherin level, and antagonized PTX-induced increase of C-C chemokine ligand 2 (CCL-2) levels by GA. Meanwhile, the synergistic anti-tumor effect of GA and PTX has also been verified in vitro and in vivo experiments. Conclusion The subcellular organelle targeted peptide modified PTX+GA multifunctional nano-drug delivery system has a good therapeutic effect on tumors, and this study provides significant insights into the role of different subcellular organelles in inhibiting tumor growth and metastasis and inspires researchers to develop highly effective cancer therapeutic strategies through subcellular organelle targeted drugs.
Collapse
Affiliation(s)
- Wenli Dang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Bin Xing
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Xintao Jia
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Ying Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Bei Jia
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Changxiang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Jiachen He
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Ziwei Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Huihui Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| | - Zhidong Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, People’s Republic of China
| |
Collapse
|
45
|
Erfani A, Schieferstein JM, Reichert P, Narasimhan CN, Pastuskovas C, Parab V, Simmons D, Yang X, Shanker A, Hammond P, Doyle PS. Crystalline Antibody-Laden Alginate Particles: A Platform for Enabling High Concentration Subcutaneous Delivery of Antibodies. Adv Healthc Mater 2023; 12:e2202370. [PMID: 36745878 PMCID: PMC11469019 DOI: 10.1002/adhm.202202370] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/30/2023] [Indexed: 02/08/2023]
Abstract
Subcutaneous (SC) administration is a desired route for monoclonal antibodies (mAbs). However, formulating mAbs for small injection volumes at high concentrations with suitable stability and injectability is a significant challenge. Here, this work presents a platform technology that combines the stability of crystalline antibodies with injectability and tunability of soft hydrogel particles. Composite alginate hydrogel particles are generated via a gentle centrifugal encapsulation process which avoids use of chemical reactions or an external organic phase. Crystalline suspension of anti-programmed cell death protein 1 (PD-1) antibody (pembrolizumab) is utilized as a model therapeutic antibody. Crystalline forms of the mAb encapsuled in the hydrogel particles lead to stable, high concentration, and injectable formulations. Formulation concentrations as high as 315 mg mL-1 antibody are achieved with encapsulation efficiencies in the range of 89-97%, with no perceivable increase in the number of antibody aggregates. Bioanalytical studies confirm superior maintained quality of the antibody in comparison with formulation approaches involving organic phases and chemical reactions. This work illustrates tuning the alginate particles' disintegration by using partially oxide alginates. Crystalline mAb-laden particles are evaluated for their biocompatibility using cell-based in vitro assays. Furthermore, the pharmacokinetics (PK) of the subcutaneously delivered human anti-PD-1 mAb in crystalline antibody-laden alginate hydrogel particles in Wistar rats is evaluated.
Collapse
Affiliation(s)
- Amir Erfani
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02142USA
| | | | | | | | | | | | | | - Xiaoyu Yang
- Merck Research LaboratoriesKenilworthNJ07033USA
| | - Apoorv Shanker
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02142USA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Paula Hammond
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02142USA
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Patrick S. Doyle
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02142USA
- Harvard Medical School Initiative for RNA MedicineBostonMA02215USA
| |
Collapse
|
46
|
Tian Z, Li X, Jiang D. Analysis of immunogenic cell death in atherosclerosis based on scRNA-seq and bulk RNA-seq data. Int Immunopharmacol 2023; 119:110130. [PMID: 37075670 DOI: 10.1016/j.intimp.2023.110130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 03/22/2023] [Accepted: 03/29/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND Regulated cell death plays a very important role in atherosclerosis (AS). Despite a large number of studies, there is a lack of literature on immunogenic cell death (ICD) in AS. METHOD Carotid atherosclerotic plaque single-cell RNA (scRNA) sequencing data were analyzed to define involved cells and determine their transcriptomic characteristics. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, CIBERSORT, ESTIMATE and ssGSEA (Gene Set Enrichment Analysis), consensus clustering analysis, random forest (RF), Decision Curve Analysis (DCA), and the Drug-Gene Interaction and DrugBank databases were applied for bulk sequencing data. All data were downloaded from Gene Expression Omnibus (GEO). RESULT mDCs and CTLs correlated obviously with AS occurrence and development (k2(mDCs) = 48.333, P < 0.001; k2(CTL) = 130.56, P < 0.001). In total, 21 differentially expressed genes were obtained for the bulk transcriptome; KEGG enrichment analysis results were similar to those for differentially expressed genes in endothelial cells. Eleven genes with a gene importance score > 1.5 were obtained in the training set and validated in the test set, resulting in 8 differentially expressed genes for ICD. A model to predict occurrence of AS and 56 drugs that may be used to treat AS were obtained with these 8 genes. CONCLUSION Immunogenic cell death occurs mainly in endothelial cells in AS. ICD maintains chronic inflammation in AS and plays a crucial role in its occurrence and development. ICD related genes may become drug-targeted genes for AS treatment.
Collapse
Affiliation(s)
- Zemin Tian
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xinyang Li
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Delong Jiang
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
47
|
Ding N, He K, Tian H, Li L, Li Q, Lu S, Ding K, Liu J, Nice EC, Zhang W, Huang C, Tang Y, Shen Z. Carrier-free delivery of thymopentin-regulated injectable nanogels via an enhanced cancer immunity cycle against melanoma metastasis. Mater Today Bio 2023; 20:100645. [PMID: 37206879 PMCID: PMC10189275 DOI: 10.1016/j.mtbio.2023.100645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023] Open
Abstract
Thymopentin (TP5), a clinically used immunomodulatory pentapeptide, can efficiently promote thymocyte differentiation and influence mature T-cell function, thus playing an essential role in the cancer immunotherapy. However, the excellent water solubility and high IC50 of TP5 result in an uncontrolled release behavior, requiring a high loading efficiency to achieve high dosage. Here in, we reported that TP5, combined with specific chemotherapeutic agents, can co-assemble into nanogels due to multiple hydrogen bonding sites. The co-assembly of TP5 with chemotherapeutic agent doxorubicin (DOX) into a carrier-free and injectable chemo-immunotherapy nanogel can enhance the cancer immunity cycle against melanoma metastasis. In this study, the designed nanogel guarantees high drug loading of TP5 and DOX and ensures a site-specific and controlled release of TP5 and DOX with minimal side effects, thus addressing the bottlenecks encountered by current chemo-immunotherapy. Moreover, the released DOX can effectively induce tumor cell apoptosis and immunogenic cell death (ICD) to activate immune initiation. Meanwhile, TP5 can significantly promote the proliferation and differentiation of dendritic cells (DCs) and T lymphocytes to amplify the cancer immunity cycle. As a result, this nanogel shows excellent immunotherapeutic efficacy against melanoma metastasis, as well as an effective strategy for TP5 and DOX application.
Collapse
Affiliation(s)
- Ning Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Kai He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Hailong Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Qiong Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuaijun Lu
- Ningbo Hospital of Ningbo University 247 Renmin Road, Jiangbei District Ningbo, Zhejiang, 315020, China
| | - Ke Ding
- Clinical Genetics Laboratory, Affiliated Hospital, Chengdu University, Chengdu 610081, China
| | - Jiaqi Liu
- International School of Public Health and Whole Health, Hainan Medical University, Haikou, 571199, PR China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Wei Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Canhua Huang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yong Tang
- Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040 Ningbo, Zhejiang, China
| |
Collapse
|
48
|
Hur W, Park Y, Seo E, Son SE, Kim S, Seo H, Seong GH. Multicomponent metal-organic framework nanocomposites for tumor-responsive synergistic therapy. J Colloid Interface Sci 2023; 645:663-675. [PMID: 37167915 DOI: 10.1016/j.jcis.2023.04.161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/04/2023] [Accepted: 04/29/2023] [Indexed: 05/13/2023]
Abstract
Targeted tumor therapy through tumor microenvironment (TME)-responsive nanoplatforms is an emerging treatment strategy used to enhance tumor-specificity to selectively kill cancer cells. Here, we introduce a nanosized zeolitic imidazolate framework-8 (ZIF-8) that simultaneously contains natural glucose oxidase (GOx) and Prussian blue nanoparticles (PBNPs) to construct multi-component metal-organic framework nanocomposites (denoted as ZIF@GOx@PBNPs), which possess cascade catalytic activity selectively within the TME. Once reaching a tumor site, GOx and PBNPs inside the nanocomposites are sequentially released and participate in the cascade catalytic reaction. In weak acidic TME, GOx, which effectively catalyzes the oxidation of intratumoral glucose to hydrogen peroxide (H2O2) and gluconic acid, not only initiates starvation therapy by cutting off the nutrition source for cancer cells but also produces the reactant for sequential Fenton reaction for chemodynamic therapy. Meanwhile, PBNPs, which are released from the ZIF-8 framework dissociated by acidified pH due to the produced gluconic acid, convert the generated H2O2 into harmful radicals to melanomas. In this way, the cascade catalytic reactions of ZIF@GOx@PBNPs enhance reactive oxygen species production and cause oxidative damage to DNA in cancer cells, resulting in remarkable inhibition of tumor growth. The tumor specificity is endowed by using the biomolecules overexpressed in TME as a "switch" to initiate the first catalytic reaction by GOx. Given the significant antitumor efficiency both in vitro and in vivo, ZIF@GOx@PBNPs could be applied as a promising therapeutic platform enabling starvation/chemodynamic synergism, high therapeutic efficiency, and minimal side effects.
Collapse
Affiliation(s)
- Won Hur
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Yeongwon Park
- Department of Molecular & Life Science, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Eunbi Seo
- Department of Molecular & Life Science, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Seong Eun Son
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Seongnyeon Kim
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Hyemyung Seo
- Department of Molecular & Life Science, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea
| | - Gi Hun Seong
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 426-791, South Korea.
| |
Collapse
|
49
|
Jia Y, Shi K, Dai L, He X, Deng H, Han R, Yang F, Chu B, Liao J, Wei X, Qian Z. Gold Nanorods and Polymer Micelles Mediated Dual TLR Stimulators Delivery System CPG@Au NRs/M-R848 Regulate Macrophages Reprogramming and DC Maturation for Enhanced Photothermal Immunotherapy of Melanoma. SMALL METHODS 2023; 7:e2201087. [PMID: 36572641 DOI: 10.1002/smtd.202201087] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/30/2022] [Indexed: 05/17/2023]
Abstract
Synergistic photothermal immunotherapy has emerged as a favorable therapeutic approach to fight cancer. However, design of an effective photothermal immunotherapy system to suppress tumor growth and simultaneously inhibit tumor metastases continues to be a challenge. Here a dual toll-like receptor agonists delivery system CPG@Au NRs/m-R848 for combined photothermal immunotherapy of melanoma is developed. CPG@Au NRs/m-R848 displays strong antitumor effects by promoting maturation of dendritic cells (DCs) and reprogramming of M2 macrophages into M1 phenotype. Moreover, immunogenic cell death (ICD) induced by photothermal ablation of Au NRs could synergistically produce in situ vaccination effect with CPG ODN and R848, generating systemic and lasting antitumor immunity. It is further proved that CPG@Au NRs/m-R848 treatment inhibits tumor growth in bilateral B16F10 tumors model by eliciting CD8+ T cell response. Overall, this work suggests that this strategy hold great potential in tumor immunotherapy by regulating tumor-associated macrophage polarization, triggering DCs maturation and inducing ICD.
Collapse
Affiliation(s)
- YanPeng Jia
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, 210000, P. R. China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - LiQun Dai
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - XinLong He
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - HanZhi Deng
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - RuXia Han
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Fan Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - BingYang Chu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - JinFeng Liao
- State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - XiaWei Wei
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - ZhiYong Qian
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
50
|
Cai M, Yao Y, Yin D, Zhu R, Fu T, Kong J, Wang K, Liu J, Yao A, Ruan Y, Shi W, Zhu Q, Ni J, Yin X. Enhanced lysosomal escape of cell penetrating peptide-functionalized metal-organic frameworks for co-delivery of survivin siRNA and oridonin. J Colloid Interface Sci 2023; 646:370-380. [PMID: 37207419 DOI: 10.1016/j.jcis.2023.04.126] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023]
Abstract
In recent years, small interfering RNA (siRNA) has been widely used in the treatment of human diseases, especially tumors, and has shown great appeal. However, the clinical application of siRNA faces several challenges. Insufficient efficacy, poor bioavailability, poor stability, and lack of responsiveness to a single therapy are the main problems affecting tumor therapy. Here, we designed a cell-penetrating peptide (CPP)-modified metal organic framework nanoplatform (named PEG-CPP33@ORI@survivin siRNA@ZIF-90, PEG-CPP33@NPs) for targeted co-delivery of oridonin (ORI), a natural anti-tumor active ingredient) and survivin siRNA in vivo. This can improve the stability and bioavailability of siRNA and the efficacy of siRNA monotherapy. The high drug-loading capacity and pH-sensitive properties of zeolite imidazolides endowed the PEG-CPP33@NPs with lysosomal escape abilities. The Polyethylene glycol (PEG)-conjugated CPP (PEG-CPP33) coating significantly improved the uptake in the PEG-CPP33@NPs in vitro and in vivo. The results showed that the co-delivery of ORI and survivin siRNA greatly enhanced the anti-tumor effect of PEG-CPP33@NPs, demonstrating the synergistic effect between ORI and survivin siRNA. In summary, the novel targeted nanobiological platform loaded with ORI and survivin siRNA presented herein showed great advantages in cancer therapy, and provides an attractive strategy for the synergistic application of chemotherapy and gene therapy.
Collapse
Affiliation(s)
- Mengru Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Dongge Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Rongyue Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tingting Fu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiahui Kong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Kaixin Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jing Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Aina Yao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yidan Ruan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Wenjuan Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qian Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|