51
|
Wang W, Zhong F, Wang D, Zhao Y, Peng D, Li S, Ning Q, Tang S, Yu CY, Wei H. Dual gatekeepers-modified mesoporous organic silica nanoparticles for synergistic photothermal-chemotherapy of breast cancer. J Colloid Interface Sci 2023; 646:118-128. [PMID: 37187045 DOI: 10.1016/j.jcis.2023.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/11/2023] [Accepted: 05/04/2023] [Indexed: 05/17/2023]
Abstract
HYPOTHESIS Construction of dual gatekeepers-functionalized mesoporous organic silica nanoparticles (MONs) with both physical and chemical mechanisms for modulated drug delivery properties provides one solution to the extracellular stability vs. intracellular high therapeutic efficiency of MONs that hold great potential for clinical translations. EXPERIMENTS We reported herein facile construction of diselenium-bridged MONs decorated with dual gatekeepers, i.e., azobenzene (Azo)/polydopamine (PDA) for both physical and chemical modulated drug delivery properties. Specifically, Azo can act as a physical barrier to block DOX in the mesoporous structure of MONs for extracellular safe encapsulation. The PDA outer corona serves not only as a chemical barrier with acidic pH-modulated permeability for double insurance of minimized DOX leakage in the extracellular blood circulation but also for inducing a PTT effect for synergistic PTT and chemotherapy of breast cancer. FINDINGS An optimized formulation, DOX@(MONs-Azo3)@PDA resulted in approximately 1.5 and 2.4 fold lower IC50 values than DOX@(MONs-Azo3) and (MONs-Azo3)@PDA controls in MCF-7 cells, respectively, and further mediated complete tumor eradication in 4T1 tumor-bearing BALB/c mice with insignificant systematic toxicity due to the synergistic PTT and chemotherapy with enhanced therapeutic efficiency.
Collapse
Affiliation(s)
- Wei Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Fengmin Zhong
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Dun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Yuqi Zhao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Dongdong Peng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Shuang Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System (2018TP1044), School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410000, China
| | - Shengsong Tang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System (2018TP1044), School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410000, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, University of South China, Hengyang 421001, China.
| |
Collapse
|
52
|
Sun Y, Du X, Liang J, Wang D, Zheng J, Bao Z, Zhao Z, Yuan Y. A multifunctional metal-organic framework nanosystem disrupts redox homeostasis for synergistic therapy. J Colloid Interface Sci 2023; 645:607-617. [PMID: 37167910 DOI: 10.1016/j.jcis.2023.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/22/2023] [Accepted: 05/01/2023] [Indexed: 05/13/2023]
Abstract
Synergistic therapies of photodynamic therapy (PDT) and chemodynamic therapy (CDT) via metal-organic frameworks (MOF) for cancer treatment have recently attracted a lot of attentions because of the limitations of insufficient reactive oxygen species (ROS) in single-modality approaches. However, few studies explored on the use of increased ROS synergized with chemotherapy (CT) to address the issue of inadequate anti-tumor efficacy in single-modality regimens. Here, the desired cascade nanoplatforms (noted as MOF(Cu)@Dox-PL NPs) were fabricated by a solvothermal method using tetrakis (4-carboxyphenyl) porphyrin (TCPP) and zirconyl(di)chloride octahydrate (ZrOCl2·8H2O) as raw material, followed by Cu2+ introduced into the porphyrin ring and doxorubicin (DOX) loaded into the nanoframework. In addition, the nanoparticles (NPs) were electrostatically and hydrophobically coated with phospholipid (PL) to improve the biocompatibility of the nanosystems. Singlet oxygen (1O2) was created by the MOF(Cu)@Dox-PL NPs to disturb intracellular redox equilibrium. The acidic microenvironment in cancer cells may cause the prior release of DOX, which encourages the production of hydrogen peroxide (H2O2). And the doped Cu2+ could deplete overexpressed reduced glutathione (GSH) to produce hydroxyl radicals (·OH) by catalyzing H2O2, further causing redox dyshomeostasis. In vivo experiments revealed that MOF(Cu)@Dox-PL nanosystem possessed good biosafety and a compelling therapeutic effect in 4T1 tumor-bearing mice. As a novel nanosystem, MOF(Cu)@Dox-PL NPs showed great potential in synergistic therapy based on redox dyshomeostasis for improving anti-tumor efficacy with high specificity.
Collapse
Affiliation(s)
- Yaning Sun
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Xuening Du
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Jingyi Liang
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Da Wang
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Jiani Zheng
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Zhihong Bao
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Zan Zhao
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | - Yue Yuan
- School of Pharmacy, Shenyang Key Laboratory of Functional Drug Carrier Materials, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China.
| |
Collapse
|
53
|
Xu X, Liu A, Liu S, Ma Y, Zhang X, Zhang M, Zhao J, Sun S, Sun X. Application of molecular dynamics simulation in self-assembled cancer nanomedicine. Biomater Res 2023; 27:39. [PMID: 37143168 PMCID: PMC10161522 DOI: 10.1186/s40824-023-00386-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Self-assembled nanomedicine holds great potential in cancer theragnostic. The structures and dynamics of nanomedicine can be affected by a variety of non-covalent interactions, so it is essential to ensure the self-assembly process at atomic level. Molecular dynamics (MD) simulation is a key technology to link microcosm and macroscale. Along with the rapid development of computational power and simulation methods, scientists could simulate the specific process of intermolecular interactions. Thus, some experimental observations could be explained at microscopic level and the nanomedicine synthesis process would have traces to follow. This review not only outlines the concept, basic principle, and the parameter setting of MD simulation, but also highlights the recent progress in MD simulation for self-assembled cancer nanomedicine. In addition, the physicochemical parameters of self-assembly structure and interaction between various assembled molecules under MD simulation are also discussed. Therefore, this review will help advanced and novice researchers to quickly zoom in on fundamental information and gather some thought-provoking ideas to advance this subfield of self-assembled cancer nanomedicine.
Collapse
Affiliation(s)
- Xueli Xu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Ao Liu
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shuangqing Liu
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Yanling Ma
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Xinyu Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Meng Zhang
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China
| | - Jinhua Zhao
- School of Science, Shandong Jianzhu University, Jinan, 250101, China
| | - Shuo Sun
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, 02115, USA
| | - Xiao Sun
- School of Chemistry and Pharmaceutical Engineering, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250000, China.
| |
Collapse
|
54
|
Jung E, Kwon S, Song N, Kim N, Jo H, Yang M, Park S, Kim C, Lee D. Tumor-targeted redox-regulating and antiangiogenic phototherapeutics nanoassemblies for self-boosting phototherapy. Biomaterials 2023; 298:122127. [PMID: 37086554 DOI: 10.1016/j.biomaterials.2023.122127] [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: 11/05/2022] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/24/2023]
Abstract
Cancer cells are equipped with abundant antioxidants such as glutathione (GSH) that eliminate reactive oxygen species (ROS) to deteriorate the therapeutic efficacy of photodynamic therapy (PDT). Another challenge in PDT is circumventing PDT-induced hypoxic condition that provokes upregulation of pro-angiogenic factor such as vascular endothelial growth factor (VEGF). It is therefore reasonable to expect that therapeutic outcomes of PDT could be maximized by concurrent delivery of photosensitizers with GSH depleting agents and VEGF suppressors. To achieve cooperative therapeutic actions of PDT with in situ GSH depletion and VEGF suppression, we developed tumor targeted redox-regulating and antiangiogenic phototherapeutic nanoassemblies (tRAPs) composed of self-assembling disulfide-bridged borylbenzyl carbonate (ssBR), photosensitizer (IR780) and tumor targeting gelatin. As a framework of tRAPs, ssBR was rationally designed to form nanoconstructs that serve as photosensitizer carriers with intrinsic GSH depleting- and VEGF suppressing ability. tRAPs effectively depleted intracellular GSH to render cancer cells more vulnerable to ROS and also provoked immunogenic cell death (ICD) of cancer cells upon near infrared (NIR) laser irradiation. In mouse xenograft models, tRAPs preferentially accumulated in tumors and dramatically eradicated tumors with laser irradiation. The design rationale of tRAPs provides a simple and versatile strategy to develop self-boosting phototherapeutic agents with great potential in targeted cancer therapy.
Collapse
Affiliation(s)
- Eunkyeong Jung
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 54896, Republic of Korea; Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA
| | - Soonyoung Kwon
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 54896, Republic of Korea
| | - Nanhee Song
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 54896, Republic of Korea
| | - Nuri Kim
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 54896, Republic of Korea
| | - Hanui Jo
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 54896, Republic of Korea
| | - Manseok Yang
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 54896, Republic of Korea
| | - Sangjun Park
- Research Institute of Radiological & Medical Sciences, Korea Institute of Radiological & Medical Sciences, Nowongu, Seoul, 01812, Republic of Korea
| | - Chunho Kim
- Research Institute of Radiological & Medical Sciences, Korea Institute of Radiological & Medical Sciences, Nowongu, Seoul, 01812, Republic of Korea
| | - Dongwon Lee
- Department of Bionanotechnology and Bioconvergence Engineering, Jeonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 54896, Republic of Korea; Department of Polymer⋅Nano Science and Technology, Jeonbuk National University, Jeonju, Jeonbuk, 54896, Republic of Korea.
| |
Collapse
|
55
|
Serrano-Sandoval SN, Jiménez-Rodríguez A, Hernández-Pérez J, Chavez-Santoscoy RA, Guardado-Félix D, Antunes-Ricardo M. Selenized Chickpea Sprouts Hydrolysates as a Potential Anti-Aging Ingredient. Molecules 2023; 28:molecules28083402. [PMID: 37110634 PMCID: PMC10145560 DOI: 10.3390/molecules28083402] [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: 03/24/2023] [Revised: 03/31/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Skin aging represents a health and aesthetic problem that could result in infections and skin diseases. Bioactive peptides can potentially be used in skin aging regulation. Chickpea (Cicer arietinum L.) selenoproteins were obtained from germination with 2 mg Na2SeO3/100 g of seeds for 2 days. Alcalase, pepsin, and trypsin were used as hydrolyzers, and a membrane < 10 kDa was used to fractionate the hydrolysate. Se content, antioxidant capacity, elastase and collagen inhibition, functional stability, and preventative capacity were analyzed. Significant increases in Se content were found in germinated chickpea flour and protein related to the control. An increase of 38% in protein was observed in the selenized flour related to the control. A band (600-550 cm-1) observed in the selenized hydrolysates suggested the insertion of Se into the protein. Hydrolysates from pepsin and trypsin had the highest antioxidant potential. Se enhanced the stability of total protein and protein hydrolysates through time and increased their antioxidant capacity. Hydrolysates > 10 kDa had higher elastase and collagenase inhibition than the total protein and hydrolysates < 10 kDa. Protein hydrolysates < 10 kDa 6 h before UVA radiation had the highest inhibition of collagen degradation. Selenized protein hydrolysates showed promising antioxidant effects that could be related to skin anti-aging effects.
Collapse
Affiliation(s)
- Sayra N Serrano-Sandoval
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, NL, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, NL, Mexico
| | - Antonio Jiménez-Rodríguez
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, NL, Mexico
| | - Jesús Hernández-Pérez
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, NL, Mexico
| | | | - Daniela Guardado-Félix
- Programa Regional de Posgrado en Biotecnología, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, FCQB-UAS, AP 1354, Culiacan 80000, SIN, Mexico
| | - Marilena Antunes-Ricardo
- Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, NL, Mexico
- The Institute for Obesity Research, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Monterrey 64849, NL, Mexico
| |
Collapse
|
56
|
Tian M, Zhu Y, Guan W, Lu C. Quantitative Measurement of Drug Release Dynamics within Targeted Organelles Using Förster Resonance Energy Transfer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2206866. [PMID: 37026420 DOI: 10.1002/smll.202206866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/01/2023] [Indexed: 06/19/2023]
Abstract
Measuring the release dynamics of drug molecules after their delivery to the target organelle is critical to improve therapeutic efficacy and reduce side effects. However, it remains challenging to quantitatively monitor subcellular drug release in real time. To address the knowledge gap, a novel gemini fluorescent surfactant capable of forming mitochondria-targeted and redox-responsive nanocarriers is designed. A quantitative Förster resonance energy transfer (FRET) platform is fabricated using this mitochondria-anchored fluorescent nanocarrier as a FRET donor and fluorescent drugs as a FRET acceptor. The FRET platform enables real-time measurement of drug release from organelle-targeted nanocarriers. Moreover, the obtained drug release dynamics can evaluate the duration of drug release at the subcellular level, which established a new quantitative method for organelle-targeted drug release. This quantitative FRET platform can compensate for the absent assessment of the targeted release performances of nanocarriers, offering in-depth understanding of the drug release behaviors at the subcellular targets.
Collapse
Affiliation(s)
- Mingce Tian
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yaping Zhu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Weijiang Guan
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chao Lu
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
- Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
57
|
Wang H, Alizadeh A, Abed AM, Piranfar A, Smaisim GF, Hadrawi SK, Zekri H, Toghraie D, Hekmatifar M. Investigation of the effects of porosity and volume fraction on the atomic behavior of cancer cells and microvascular cells of 3DN5 and 5OTF macromolecular structures during hematogenous metastasis using the molecular dynamics method. Comput Biol Med 2023; 158:106832. [PMID: 37037148 DOI: 10.1016/j.compbiomed.2023.106832] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/03/2023] [Accepted: 03/26/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND AND OBJECTIVE The molecular dynamics (MD) simulation is a powerful tool for researching how cancer patients are treated. The efficiency of many factors may be predicted using this approach in great detail and with atomic accuracy. METHODS The MD simulation method was used to investigate the impact of porosity and the number of cancer cells on the atomic behavior of cancer cells during the hematogenous spread. In order to examine the stability of simulated structures, temperature and potential energy (PE) values are used. To evaluate how cell structure has changed, physical parameters such as gyration radius, interaction force, and interaction energy are also used. RESULTS The findings demonstrate that the samples' gyration radius, interaction energy, and interaction force rose from 41.33 Å, -551.38 kcal/mol, and -207.10 kcal/mol Å to 49.49, -535.94 kcal/mol, and -190.05 kcal/mol Å, respectively, when the porosity grew from 0% to 5%. Also, the interaction energy and force in the samples fell from -551.38 kcal/mol and -207.10 kcal/mol to -588.03 kcal/mol and -237.81 kcal/mol Å, and the amount of gyration radius reduced from 41.33 to 37.14 Å as the number of cancer cells rose from 1 to 5 molecules. The strength and stability of the simulated samples will improve when the radius of gyration is decreased. CONCLUSIONS Therefore, high accumulation of cancer cells will make them resistant to atomic collapse. It is expected that the results of this simulation should be used to optimize cancer treatment processes further.
Collapse
|
58
|
Ma Z, Wang H, Shi Z, Yan F, Li Q, Chen J, Cui ZK, Zhang Y, Jin X, Jia YG, Wang L. Inhalable GSH-Triggered Nanoparticles to Treat Commensal Bacterial Infection in In Situ Lung Tumors. ACS NANO 2023; 17:5740-5756. [PMID: 36884352 DOI: 10.1021/acsnano.2c12165] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Bacterial infection has been considered one of the primary reasons for low survival rate of lung cancer patients. Herein, we demonstrated that a kind of mesoporous silica nanoparticles loaded with anticancer drug doxorubicin (DOX) and antimicrobial peptide HHC36 (AMP) (MSN@DOX-AMP) can kill both commensal bacteria and tumor cells under GSH-triggering, modulating the immunosuppressive tumor microenvironment, significantly treating commensal bacterial infection, and eliminating in situ lung tumors in a commensal model. Meanwhile, MSN@DOX-AMP encapsulated DOX and AMP highly efficiently via a combined strategy of physical adsorption and click chemistry and exhibited excellent hemocompatibility and biocompatibility. Importantly, MSN@DOX-AMP could be inhaled and accumulate in lung by a needle-free nebulization, achieving a better therapeutic effect. This system is expected to serve as a straightforward platform to treat commensal bacterial infections in tumors and promote the translation of such inhaled GSH-triggered MSN@DOX-AMP to clinical treatments of lung cancer.
Collapse
Affiliation(s)
- Zunwei Ma
- School of Materials Science & Engineering, South China University of Technology, Guangzhou 510006, China
| | - Huaiming Wang
- Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510655, China
| | - Zhifeng Shi
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Fengying Yan
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Qingtao Li
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Junjian Chen
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Zhong-Kai Cui
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yunjiao Zhang
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xin Jin
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Yong-Guang Jia
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Lin Wang
- School of Materials Science & Engineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
59
|
Park JE, Lee H, Oliva P, Kirsch K, Kim B, Ahn JI, Alverez CN, Gaikwad S, Krausz KW, O’Connor R, Rai G, Simeonov A, Mock BA, Gonzalez FJ, Lee KS, Jacobson KA. Structural Optimization and Anticancer Activity of Polo-like Kinase 1 (Plk1) Polo-Box Domain (PBD) Inhibitors and Their Prodrugs. ACS Pharmacol Transl Sci 2023; 6:422-446. [PMID: 36926457 PMCID: PMC10012257 DOI: 10.1021/acsptsci.2c00250] [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: 12/23/2022] [Indexed: 02/22/2023]
Abstract
Polo-like kinase 1 (Plk1), a mitotic kinase whose activity is widely upregulated in various human cancers, is considered an attractive target for anticancer drug discovery. Aside from the kinase domain, the C-terminal noncatalytic polo-box domain (PBD), which mediates the interaction with the enzyme's binding targets or substrates, has emerged as an alternative target for developing a new class of inhibitors. Various reported small molecule PBD inhibitors exhibit poor cellular efficacy and/or selectivity. Here, we report structure-activity relationship (SAR) studies on triazoloquinazolinone-derived inhibitors, such as 43 (a 1-thioxo-2,4-dihydrothieno[2,3-e][1,2,4]triazolo[4,3-a]pyrimidin-5(1H)-one) that effectively block Plk1, but not Plk2 and Plk3 PBDs, with improved affinity and drug-like properties. The range of prodrug moieties needed for thiol group masking of the active drugs has been expanded to increase cell permeability and mechanism-based cancer cell (L363 and HeLa) death. For example, a 5-thio-1-methyl-4-nitroimidazolyl prodrug 80, derived from 43, showed an improved cellular potency (GI50 4.1 μM). As expected, 80 effectively blocked Plk1 from localizing to centrosomes and kinetochores and consequently induced potent mitotic block and apoptotic cell death. Another prodrug 78 containing 9-fluorophenyl in place of the thiophene-containing heterocycle in 80 also induced a comparable degree of anti-Plk1 PBD effect. However, orally administered 78 was rapidly converted in the bloodstream to parent drug 15, which was shown be relatively stable toward in vivo oxidation due to its 9-fluorophenyl group in comparison to unsubstituted phenyl. Further derivatization of these inhibitors, particularly to improve the systemic prodrug stability, could lead to a new class of therapeutics against Plk1-addicted cancers.
Collapse
Affiliation(s)
- Jung-Eun Park
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hobin Lee
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Laboratory
of Bioorganic Chemistry, National Institute
of Diabetes and Digestive and Kidney Diseases, National Institutes
of Health, Bethesda, Maryland 20892, United States
| | - Paola Oliva
- Laboratory
of Bioorganic Chemistry, National Institute
of Diabetes and Digestive and Kidney Diseases, National Institutes
of Health, Bethesda, Maryland 20892, United States
| | - Klara Kirsch
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Bora Kim
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Jong Il Ahn
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Celeste N. Alverez
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
- Division
of Preclinical Innovation, National Center
for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Snehal Gaikwad
- Laboratory
of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of
Health, Bethesda, Maryland 20892, United States
| | - Kristopher W. Krausz
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Robert O’Connor
- Laboratory
of Bioorganic Chemistry, National Institute
of Diabetes and Digestive and Kidney Diseases, National Institutes
of Health, Bethesda, Maryland 20892, United States
| | - Ganesha Rai
- Division
of Preclinical Innovation, National Center
for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Anton Simeonov
- Division
of Preclinical Innovation, National Center
for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850, United States
| | - Beverly A. Mock
- Laboratory
of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of
Health, Bethesda, Maryland 20892, United States
| | - Frank J. Gonzalez
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kyung S. Lee
- Cancer
Innovation Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Kenneth A. Jacobson
- Laboratory
of Bioorganic Chemistry, National Institute
of Diabetes and Digestive and Kidney Diseases, National Institutes
of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
60
|
Sun T, Jiang C. Stimuli-responsive drug delivery systems triggered by intracellular or subcellular microenvironments. Adv Drug Deliv Rev 2023; 196:114773. [PMID: 36906230 DOI: 10.1016/j.addr.2023.114773] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023]
Abstract
Drug delivery systems (DDS) triggered by local microenvironment represents the state-of-art of nanomedicine design, where the triggering hallmarks at intracellular and subcellular levels could be employed to exquisitely recognize the diseased sites, reduce side effects, and expand the therapeutic window by precisely tailoring the drug-release kinetics. Though with impressive progress, the DDS design functioning at microcosmic levels is fully challenging and underexploited. Here, we provide an overview describing the recent advances on stimuli-responsive DDSs triggered by intracellular or subcellular microenvironments. Instead of focusing on the targeting strategies as listed in previous reviews, we herein mainly highlight the concept, design, preparation and applications of stimuli-responsive systems in intracellular models. Hopefully, this review could give useful hints in developing nanoplatforms proceeding at a cellular level.
Collapse
Affiliation(s)
- Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, PR China.
| |
Collapse
|
61
|
Diselenide-triggered hydroxyethyl starch conjugate nanoparticles with cascade drug release properties for potentiating chemo-photodynamic therapy. Carbohydr Polym 2023; 311:120748. [PMID: 37028875 DOI: 10.1016/j.carbpol.2023.120748] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023]
Abstract
A novel type of diselenide bond-bridged hydroxyethyl starch-doxorubicin conjugate, HES-SeSe-DOX, was synthesized via a specially designed multistep synthetic route. The optimally achieved HES-SeSe-DOX was further combined with photosensitizer, chlorin E6 (Ce6), to self-assemble into HES-SeSe-DOX/Ce6 nanoparticles (NPs) for potentiating chemo-photodynamic anti-tumor therapy via diselenide-triggered cascade actions. HES-SeSe-DOX/Ce6 NPs were observed to disintegrate through the cleavage or oxidation of diselenide-bridged linkages in response to the stimuli arising from glutathione (GSH), hydrogen peroxide and Ce6-induced singlet oxygen, respectively, as evidenced by the enlarged size with irregular shapes and cascade drug release. In vitro cell studies exhibited that HES-SeSe-DOX/Ce6 NPs in combination with laser irradiation effectively consumed intracellular GSH and promoted a large rise in levels of reactive oxygen species in tumor cells, actuating the disruption of intracellular redox balance and the enhanced chemo-photodynamic cytotoxicity against tumor cells. The in vivo investigations revealed that HES-SeSe-DOX/Ce6 NPs were inclined to accumulate in tumors with persistent fluorescence emission, inhibited tumor growth with high efficacy and had good safety. These findings demonstrate the potential of HES-SeSe-DOX/Ce6 NPs for use in chemo-photodynamic tumor therapy and suggest their viability for clinical translation.
Collapse
|
62
|
Synthesis and Application Dichalcogenides as Radical Reagents with Photochemical Technology. Molecules 2023; 28:molecules28041998. [PMID: 36838986 PMCID: PMC9963440 DOI: 10.3390/molecules28041998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/23/2023] Open
Abstract
Dichalcogenides (disulfides and diselenides), as reactants for organic transformations, are important and widely used because of their potential to react with nucleophiles, electrophilic reagents, and radical precursors. In recent years, in combination with photochemical technology, the application of dichalcogenides as stable radical reagents has opened up a new route to the synthesis of various sulfur- and selenium-containing compounds. In this paper, synthetic strategies for disulfides and diselenides and their applications with photochemical technology are reviewed: (i) Cyclization of dichalcogenides with alkenes and alkynes; (ii) direct selenylation/sulfuration of C-H/C-C/C-N bonds; (iii) visible-light-enabled seleno- and sulfur-bifunctionalization of alkenes/alkynes; and (iv) Direct construction of the C(sp)-S bond. In addition, the scopes, limitations, and mechanisms of some reactions are also described.
Collapse
|
63
|
Yan D, Wu Z, Qi X. Ferroptosis-Related Metabolic Mechanism and Nanoparticulate Anticancer Drug Delivery Systems Based on Ferroptosis. Saudi Pharm J 2023; 31:554-568. [PMID: 37063438 PMCID: PMC10102556 DOI: 10.1016/j.jsps.2023.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Ferroptosis is a new type of cell death discovered in recent years that distinguishes from apoptosis and necrosis, mainly caused by the imbalance between the production and degradation of lipid reactive oxygen species in cells. Although the mechanism of ferroptosis is not yet clear, the phenomenon of ferroptosis has attracted widespread attention from researchers and has become a new hotspot in anti-tumor research. Studies have shown that ferroptosis is involved in the occurrence and development of a variety of diseases such as nervous system diseases, cardiovascular diseases and cancer. And inhibiting or inducing the occurrence of ferroptosis can effectively intervene in related diseases. At the same time, nanotechnology, by virtue of its distinct advantages, has been widely used in the development of nanodrug delivery systems. This review outlines current the advance on the intersection of ferroptosis and biomedical nanotechnology. In this review, the discovery and characteristics of ferroptosis, the mechanism of occurrence and the relationship with disease are summarized. More importantly, we summarized the strategies for inducing ferroptosis based on nanoparticulate drug delivery systems for cancer treatment.
Collapse
|
64
|
Sun Y, Wang S, Li Y, Wang D, Zhang Y, Zhang H, Lei H, Liu X, Sun J, Sun B, He Z. Precise engineering of disulfide bond-bridged prodrug nanoassemblies to balance antitumor efficacy and safety. Acta Biomater 2023; 157:417-427. [PMID: 36513247 DOI: 10.1016/j.actbio.2022.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
Prodrug-based nanoassemblies, which combine the merits of prodrug technology and nanocarriers, are regarded as promising platforms for cancer treatment. Notably, the chemical structure of prodrugs is closely associated with antitumor efficacy and safety, and the intrinsic relationships among them need further exploration. Herein, paclitaxel was conjugated with 2-octyldodecan-1-ol through different positions of disulfide bond to construct the prodrug nanoassemblies. Interestingly, the minor differences in chemical structure not only dominated the assembly performance and drug release of nanoassemblies, but also significantly impacted the pharmacokinetics, antitumor efficacy, and safety. It was worth noting that prodrug nanoassemblies with one carbon atom between disulfide bond and ester bond had faster drug release and better antitumor effect, while prodrug nanoassemblies with three carbon atoms between disulfide bond and ester bond possessed moderate antitumor effect and better safety. Our findings illustrated the structure-function relationships of self-assembled prodrugs and provided a promising paradigm for the precise engineering of advanced prodrug nanoplatforms. STATEMENT OF SIGNIFICANCE: 1. The major effects of minor differences in prodrug chemical structure on pharmacodynamics and safety were explored, which had important clinical reference significance and value. 2. The in-depth exploration of structure-function relationships to balance efficacy and safety had important guiding significance for the design of prodrug nanoassemblies.
Collapse
Affiliation(s)
- Yixin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Simeng Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yaqi Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Danping Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hongrui Lei
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaohong Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
65
|
Wang X, Liu T, Huang Y, Dong F, Li L, Song J, Zuo S, Zhu Z, Kamei KI, He Z, Sun B, Sun J. Critical roles of linker length in determining the chemical and self-assembly stability of SN38 homodimeric nanoprodrugs. NANOSCALE HORIZONS 2023; 8:235-244. [PMID: 36537183 DOI: 10.1039/d2nh00425a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Homodimeric prodrug nanoassemblies (HDPNs) have been widely studied for efficient cancer therapy by virtue of their ultra-high drug loading and distinct nanostructure. However, the development of SN38 HDPNs is still a great challenge due to the rigid planar aromatic ring structure. Improving the structural flexibility of homodimeric prodrugs by increasing the linker length may be a potential strategy for constructing SN38 HDPNs. Herein, three SN38 homodimeric prodrugs with different linker lengths were synthesized. The number of carbon atoms from the disulfide bond to the adjacent ester bond is 1 (denoted as α-SN38-SS-SN38), 2 (β-SN38-SS-SN38), and 3 (γ-SN38-SS-SN38), respectively. Interestingly, we found that α-SN38-SS-SN38 exhibited extremely low yield and poor chemical stability. Additionally, β-SN38-SS-SN38 demonstrated suitable chemical stability but poor self-assembly stability. In comparison, γ-SN38-SS-SN38 possessed good chemical and self-assembly stability, thereby improving the tumor accumulation and antitumor efficacy of SN38. We developed the SN38 HDPNs for the first time and illustrated the underlying molecular mechanism of increasing the linker length to enhance the chemical and self-assembly stability of homodimeric prodrugs. These findings would provide new insights for the rational design of HDPNs with superior performance.
Collapse
Affiliation(s)
- Xin Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
- Department of Radiology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, P. R. China
| | - Tian Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Yuetong Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Fudan Dong
- Henan Provincial People's Hospital, Zhengzhou, 450003, P. R. China
| | - Lingxiao Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Jiaxuan Song
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Shiyi Zuo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Zhengyang Zhu
- Department of Radiology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, P. R. China
| | - Ken-Ichiro Kamei
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Bingjun Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, P. R. China
| |
Collapse
|
66
|
Reprogramming systemic and local immune function to empower immunotherapy against glioblastoma. Nat Commun 2023; 14:435. [PMID: 36702831 PMCID: PMC9880004 DOI: 10.1038/s41467-023-35957-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
The limited benefits of immunotherapy against glioblastoma (GBM) is closely related to the paucity of T cells in brain tumor bed. Both systemic and local immunosuppression contribute to the deficiency of tumor-infiltrating T cells. However, the current studies focus heavily on the local immunosuppressive tumor microenvironment but not on the co-existence of systemic immunosuppression. Here, we develop a nanostructure named Nano-reshaper to co-encapsulate lymphopenia alleviating agent cannabidiol and lymphocyte recruiting cytokine LIGHT. The results show that Nano-reshaper increases the number of systemic T cells and improves local T-cell recruitment condition, thus greatly increasing T-cell infiltration. When combined with immune checkpoint inhibitor, this therapeutic modality achieves 83.3% long-term survivors without recurrence in GBM models in male mice. Collectively, this work unveils that simultaneous reprogramming of systemic and local immune function is critical for T-cell based immunotherapy and provides a clinically translatable option for combating brain tumors.
Collapse
|
67
|
Zhao X, Guo H, Bera H, Jiang H, Chen Y, Guo X, Tian X, Cun D, Yang M. Engineering Transferrin-Decorated Pullulan-Based Prodrug Nanoparticles for Redox Responsive Paclitaxel Delivery to Metastatic Lung Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:4441-4457. [PMID: 36633929 DOI: 10.1021/acsami.2c18422] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Paclitaxel (PTX) remains a cornerstone in the treatment of locally advanced and metastatic lung cancer. To improve its therapeutic indices against lung cancer, novel redox-sensitive pullulan/PTX-based prodrug NPs (PULL-SS-PTX NPs) were accomplished, which were further surface-decorated with transferrin (TF), a cancer cell-targeting ligand, to afford TF-PULL-SS-PTX NPs. These prodrug NPs (drug content, >37% and average size, 134-163 nm) rapidly dismantled their self-assembled architecture upon exposure to simulated reducing conditions, causing a triggered drug release as compared to the control scaffold (PULL-CC-PTX NPs). These scaffolds also evidenced outstanding colloidal stability, cellular uptake efficiency, and discriminating cytotoxicity between the cancer and healthy cells. Intravenously delivered redox-sensitive NPs exhibited improved tumor-suppressing properties as compared to the control nanovesicles (PULL-CC-PTX NPs) in a B16-F10 melanoma lung metastasis mice model. The targeting efficiency and associated augmented anticancer potentials of TF-PULL-SS-PTX NPs relative to TF-free redox-responsive NPs and Taxol intravenous injection were also established on the transferrin receptor (TFR) overexpressed Lewis lung carcinoma (LLC-luc) cell-bearing mice model. Moreover, the TF-functionalized scaffold displayed a reduced systemic toxicity compared to that of Taxol intravenous injection. Overall, the proposed TF-decorated prodrug NPs could be a promising nanomedicine for intracellular PTX delivery against metastatic lung cancer.
Collapse
Affiliation(s)
- Xing Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
| | - Haifei Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
| | - Hriday Bera
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
- Dr. B. C. Roy College of Pharmacy and Allied Health Sciences, Dr. Meghnad Saha Sarani, Durgapur, India713206
| | - Huiyang Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
| | - Yang Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
| | - Xiong Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
| | - Xidong Tian
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road No. 103, 110016Shenyang, China
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100Copenhagen, Denmark
| |
Collapse
|
68
|
Liu Y, Wang X, Wang Z, Liao R, Qiu Q, Wang Y, Luo C. Reduction-Responsive Stearyl Alcohol-Cabazitaxel Prodrug Nanoassemblies for Cancer Chemotherapy. Pharmaceutics 2023; 15:262. [PMID: 36678891 PMCID: PMC9864162 DOI: 10.3390/pharmaceutics15010262] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Cabazitaxel (CTX) has distinct therapeutic merits for advanced and metastatic cancer. However, the present clinical formulation (Jevtana®) has several defects, especially for undesirable tumor-targeting and serious side effects, greatly limiting the therapeutic efficacy. Small-molecule prodrug-based nanoassemblies integrate the advantages of both prodrug strategy and nanotechnology, emerging as a promising treatment modality. Herein, disulfide bonds with different lengths were employed as linkages to elaborately synthesize three redox-sensitive stearyl alcohol (SAT)-CTX prodrug-based nanoassemblies (SAC NPs, SBC NPs and SGC NPs) for seeking optimal chemotherapeutical treatment. All the prodrug-based nanoassemblies exhibited impressive drug-loading efficiency, superior self-assembly capability and excellent colloidal stability. Interestingly, the drug release behaviors of three prodrug-nanoassemblies in the same reductive environment were different owing to tiny changes in the carbon chain length of disulfide bonds, resulting in disparate cytotoxicity effects, pharmacokinetic outcomes and in vivo antitumor efficacies. Among them, SAC NPs displayed rapid drug release, excellent cytotoxicity, long blood circulation and enhanced tumor accumulation, thus showing strong tumor inhibition in the 4T1-bearing mouse model. Our study shed light on the vital role of connecting bonds in designing high-efficiency, low-toxicity prodrug nanoassemblies.
Collapse
Affiliation(s)
| | | | | | | | | | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
69
|
Wang Y, Xia H, Chen B, Wang Y. Rethinking nanoparticulate polymer-drug conjugates for cancer theranostics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1828. [PMID: 35734967 DOI: 10.1002/wnan.1828] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 01/31/2023]
Abstract
Polymer-drug conjugates (PDCs) fabricated as nanoparticles have hogged the limelight in cancer theranostics in the past decade. Many researchers have devoted to developing novel and efficient polymeric drug delivery system since the first generation of poly(N-[2-hydroxypropyl]methacrylamide) copolymer-drug conjugates. However, none of them has been approved for chemotherapy in clinic. An ideal PDC nanoparticle for cancer theranostics should possess several properties, including prolonged circulation in blood, sufficient accumulation and internalization in tumors, and efficient drug release in target sites. To achieve these goals, it is important to rationally design the nanoparticulate PDCs based on circulation, accumulation, penetration, internalization, and drug release (CAPIR) cascade. Specifically, CAPIR cascades are divided into five steps: (1) circulation in the vascular compartment without burst release, (2) accumulation in tumors via enhanced permeability and retention effect, (3) subsequent penetration into the deep regions of tumors, (4) internalization into tumor cells, and (5) release of drugs as free molecules to exert their pharmacological effects. In this review, we focus on the development and novel approaches of nanoparticulate PDCs based on CAPIR cascade, and provide an outlook on future clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yaoqi Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China.,Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
| | - Heming Xia
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
70
|
Ma X, Wang P, Wu Q, Zhou J, Wang D, Yadav D, Zhang H, Zhang Y. Porphyrin Centered Paclitaxel Tetrameric Prodrug Nanoassemblies as Tumor-Selective Theranostics for Synergized Breast Cancer Therapy. Adv Healthc Mater 2023; 12:e2202024. [PMID: 36222266 DOI: 10.1002/adhm.202202024] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/25/2022] [Indexed: 01/18/2023]
Abstract
Although having undergone decades of development, nanoparticulate drug delivery vehicles for efficient cancer therapy remain a challenge, confined by low drug loading, instability, and poor cancer tissue selectivity. A self-assembled prodrug, the combination of prodrug strategy and the self-assembly merits, represents a special chemical entity which spontaneously organizes into supramolecular composites with defined architecture, therefore also providing a strategy to develop new medications. Paclitaxel (PTX) is still among the most generally prescribed chemotherapeutics in oncology but is restricted by poor solubility. Although photodynamic therapy, with its noninvasive features and barely developed drug resistance, signifies an alternative tool to suppress life-threatening cancer, sole use hardly fulfills its potential. To this end, a reduction-activatable heterotetrameric prodrug with the photosensitizer is synthesized, then formulated into self-assembled nanoparticles (NPs) for tumor imaging and combined chemo- and photodynamic therapy. Coating the NPs with amphiphilic polymer distearylphosphatidylethanolamine-polyethylene glycol-arginine-glycine-aspartate (DSPE-PEG-RGD) offers high stability and enables cancer tissue targeting. The as-prepared NPs enlighten disease cells and reveal more potent cytotoxicity comparing to PTX and the photosensitizer alone. Furthermore, the NPs selectively accumulates into tumors and synergistically inhibits tumor proliferation with reduced side effects in mice.
Collapse
Affiliation(s)
- Xiaodong Ma
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China.,Ningbo Institute of Northwestern Polytechnical University, Frontiers Science Center for Flexible Electronics (FSCFE), Key laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China.,Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Helsinki, FI-00520, Finland
| | - Pengfei Wang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China.,Ningbo Institute of Northwestern Polytechnical University, Frontiers Science Center for Flexible Electronics (FSCFE), Key laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| | - Qiwei Wu
- Department of Radiology Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Junnian Zhou
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Helsinki, FI-00520, Finland
| | - Dongqing Wang
- Department of Radiology Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Deependra Yadav
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China.,Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Helsinki, FI-00520, Finland
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Helsinki, FI-00520, Finland
| | - Yuezhou Zhang
- Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, China.,Ningbo Institute of Northwestern Polytechnical University, Frontiers Science Center for Flexible Electronics (FSCFE), Key laboratory of Flexible Electronics of Zhejiang Province, Ningbo Institute of Northwestern Polytechnical University, 218 Qingyi Road, Ningbo, 315103, China
| |
Collapse
|
71
|
Jiang Q, Lu S, Xu X, Bai C, Yan Q, Fang M, Huang L, Jin C, Zhang Y, Sun J, He Z, Zhao C, Qin F, Wang Y, Zhang T. Inhibition of alanine-serine-cysteine transporter 2-mediated auto-enhanced photodynamic cancer therapy of co-nanoassembly between V-9302 and photosensitizer. J Colloid Interface Sci 2023; 629:773-784. [PMID: 36195017 DOI: 10.1016/j.jcis.2022.05.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/28/2022] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
The efficiency of reactive oxygen species (ROS)-based photodynamic therapy (PDT) is far from satisfactory, because cancer cells can adapt to PDT by upregulating glutathione (GSH) levels. The GSH levels in tumor cells are determined based on glutamine availability via alanine-serine-cysteine transporter 2 (ASCT2)-mediated entry into cells. Herein, we develop co-assembled nanoparticles (PPa/V-9302 NPs) of the photosensitizer pyropheophorbide a (PPa) and V-9302 (a known inhibitor of ASCT2) in a 1:1 M ratio using a one-step precipitation method to auto-enhance photodynamic therapy. The computational simulations revealed that PPa and V-9302 could self-assemble through different driving forces, such as π-π stacking, hydrophobic interactions, and ionic bonds. Such PPa/V-9302 NPs could disrupt the intracellular redox homeostasis due to enhanced ROS production via PPa-induced PDT and reduced GSH synthesis via inhibition of the ASCT2-mediated glutamine flux by V-9302. The in vivo assays reveal that PPa/V-9302 NPs could increase the drug accumulation in tumor sites and suppress tumor growth in BALB/c mice bearing mouse breast carcinoma (4 T1) tumor. Our findings provide a new paradigm for the rational design of the PDT-based combinational cancer therapy.
Collapse
Affiliation(s)
- Qikun Jiang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Sirun Lu
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaolan Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chenxia Bai
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qing Yan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengna Fang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Li Huang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | | | - Yunran Zhang
- Shanghai Institute of Pharmacists, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai 315615, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chunyang Zhao
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110002, China.
| | - Feng Qin
- Department of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yongjun Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Tianhong Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
72
|
Tailoring carrier-free nanocombo of small-molecule prodrug for combinational cancer therapy. J Control Release 2022; 352:256-275. [PMID: 36272660 DOI: 10.1016/j.jconrel.2022.10.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
The outcomes of monotherapy could not satisfy clinical cancer treatment owing to the challenges of tumor heterogeneity, multi-drug resistance, tumor metastasis and relapse. In response, the significance of combinational cancer therapy has been highlighted. Traditional combinational schemes usually utilize "free" drug for multi drug administration, independently. The diverse pharmacokinetics and biodistribution greatly hinder the antitumor effects and cause systematic toxicity. To tackle the hinderance, various nanoparticulate drug delivery systems (Nano-DDSs) have been developed. However, conventional Nano-DDSs encapsulate drugs into carrier materials through noncovalent interactions, resulting in low drug loading, fixed multi drug encapsulation ratio, chemical instability and carrier-associated toxicity. Recently, carrier-free nanocombos based on self-assembling small-molecule prodrugs (SPNCs) have emerged as a versatile Nano-DDSs for multiple drug delivery. Benefited by the self-assembly capability, SPNCs could be facilely fabricated with distinct merits of ultra-high drug loading, adjustable drug ratio and negligible carrier-associated toxicity. Herein, we summarize the latest trends of SPNCs. First, a basic review on self-assembling small-molecule prodrugs is presented. Additionally, facile techniques to prepare SPNCs are introduced. Furthermore, advanced combinational therapies based on SPNCs are spotlighted with special emphasis on synergistic mechanisms. Finally, future prospects and challenges are discussed.
Collapse
|
73
|
Liu T, Li L, Wang S, Dong F, Zuo S, Song J, Wang X, Lu Q, Wang H, Zhang H, Cheng M, Liu X, He Z, Sun B, Sun J. Hybrid chalcogen bonds in prodrug nanoassemblies provides dual redox-responsivity in the tumor microenvironment. Nat Commun 2022; 13:7228. [PMID: 36434014 PMCID: PMC9700694 DOI: 10.1038/s41467-022-35033-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022] Open
Abstract
Sulfur bonds, especially trisulfide bond, have been found to ameliorate the self-assembly stability of homodimeric prodrug nanoassemblies and could trigger the sensitive reduction-responsive release of active drugs. However, the antitumor efficacy of homodimeric prodrug nanoassemblies with single reduction-responsivity may be restricted due to the heterogeneous tumor redox microenvironment. Herein, we replace the middle sulfur atom of trisulfide bond with an oxidizing tellurium atom or selenium atom to construct redox dual-responsive sulfur-tellurium-sulfur and sulfur-selenium-sulfur hybrid chalcogen bonds. The hybrid chalcogen bonds, especially the sulfur-tellurium-sulfur bond, exhibit ultrahigh dual-responsivity to both oxidation and reduction conditions, which could effectively address the heterogeneous tumor microenvironment. Moreover, the hybrid sulfur-tellurium-sulfur bond promotes the self-assembly of homodimeric prodrugs by providing strong intermolecular forces and sufficient steric hindrance. The above advantages of sulfur-tellurium-sulfur bridged homodimeric prodrug nanoassemblies result in the improved antitumor efficacy of docetaxel with satisfactory safety. The exploration of hybrid chalcogen bonds in drug delivery deepened insight into the development of prodrug-based chemotherapy to address tumor redox heterogeneity, thus enriching the design theory of prodrug-based nanomedicines.
Collapse
Affiliation(s)
- Tian Liu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Lingxiao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Shuo Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Fudan Dong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Shiyi Zuo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Jiaxuan Song
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Xin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Helin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Haotian Zhang
- Department of Pharmacology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Xiaohong Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 110016, Shenyang, People's Republic of China.
| |
Collapse
|
74
|
Nguyen MP, Pham DP, Kim D. Oxidative Stress-Induced Silver Nano-Carriers for Chemotherapy. Pharmaceuticals (Basel) 2022; 15:ph15121449. [PMID: 36558899 PMCID: PMC9783686 DOI: 10.3390/ph15121449] [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: 10/10/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
Recently, silver nanoparticles (AgNPs) have been extensively explored in a variety of biological applications, especially cancer treatment. AgNPs have been demonstrated to exhibit anti-tumor effects through cell apoptosis. This study intends to promote cell apoptosis further by increasing oxidative stress. AgNPs are encapsulated by biocompatible and biodegradable polyaspartamide (PA) (PA-AgNPs) that carries the anti-cancer drug Doxorubicin (Dox) to inhibit cancer cells primarily. PA-AgNPs have an average hydrodynamic diameter of 130 nm, allowing them to move flexibly within the body. PA-AgNPs show an excellent targeting capacity to cancer cells when they are conjugated to biotin. In addition, they release Dox efficiently by up to 88% in cancer environments. The DCFDA experiment demonstrates that the Dox-carried PA-AgNPs generate reactive oxidation species intensively beside 4T1 cells. The MTT experiment confirms that PA-AgNPs with Dox may strongly inhibit 4T1 cancer cells. Furthermore, the in vivo study confirms that PA-AgNPs with Dox successfully inhibit tumors, which are about four times smaller than the control group and have high biosafety that can be applied for chemotherapy.
Collapse
Affiliation(s)
- Minh Phuong Nguyen
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Duy Phong Pham
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dukjoon Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Correspondence:
| |
Collapse
|
75
|
Xu X, Liu Y, Shao S, Li J, Xu Z, Yin Y, Zhao L, Wang Y, Liu D. Synthesis of Paclitaxel Derivatives for Remote Loading into Liposomes and Improved Therapeutic Effect. Molecules 2022; 27:molecules27227967. [PMID: 36432067 PMCID: PMC9694711 DOI: 10.3390/molecules27227967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
A series of novel paclitaxel derivatives modified by boronic acid according to the characteristics of the interaction between RB(OH)2 and different strapping agents of intraliposomal aqueous phase were designed and synthesized, which were then used to develop remote poorly water-soluble drugs loading into liposomes. Meanwhile, we screened nineteen paclitaxel boronic acid derivatives for their cytotoxic activities against three cancer cell lines (A549, HCT-116 and 4T1) and one normal cell line (LO2), and performed liposome formulation screening of active compounds. Among all the compounds, the liposome of 4d, with excellent drug-encapsulated efficiency (>95% for drug-to-lipid ratio of 0.1 w/w), was the most stable. Furthermore, the liposomes of compound 4d (8 mg/kg, 4 times) and higher dose of compound 4d (24 mg/kg, 4 times) showed better therapeutic effect than paclitaxel (8 mg/kg, 4 times) in the 4T1 tumor model in vivo, and the rates of tumor inhibition were 74.3%, 81.9% and 58.5%, respectively. This study provided a reasonable design strategy for the insoluble drugs to improve their drug loading into liposomes and anti-tumor effect in vivo.
Collapse
Affiliation(s)
- Xiangwei Xu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, China
| | - Shanshan Shao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinbo Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhaochu Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yueling Yin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Linxiang Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence: (L.Z.); (Y.W.); (D.L.); Tel.: +86-024-4352-0218 (D.L.)
| | - Yongjun Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence: (L.Z.); (Y.W.); (D.L.); Tel.: +86-024-4352-0218 (D.L.)
| | - Dan Liu
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
- Correspondence: (L.Z.); (Y.W.); (D.L.); Tel.: +86-024-4352-0218 (D.L.)
| |
Collapse
|
76
|
Wang D, Du C, Wang S, Li L, Liu T, Song J, He Z, Zhai Y, Sun B, Sun J. Probing the Role of Connecting Bonds and Modifying Chains in the Rational Design of Prodrug Nanoassemblies. ACS APPLIED MATERIALS & INTERFACES 2022; 14:51200-51211. [PMID: 36397309 DOI: 10.1021/acsami.2c14523] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Prodrug-based self-assembled nanoparticles combined with the merits of nanotechnology and prodrugs strategies have gradually become a research trending topic in the field of drug delivery. These prodrugs usually consist of parent drugs, connecting bonds, and modifying chains. The influences of the connecting bonds and modifying chains on the pharmaceutical characteristics, in vivo delivery fate, and antitumor activity of prodrug nanoassemblies remain elusive. Herein, three docetaxel (DTX) prodrugs were designed using sulfur bonds (thioether bond or disulfide bond) as connecting bonds and fatty alcohols (straight chain or branched chain) as modifying chains. Interestingly, the difference between connecting bonds and modifying chains deeply influenced the colloidal stability, redox responsive drug release, cytotoxicity, pharmacokinetic properties, tumor accumulation, and antitumor effect of prodrug nanoassemblies. DTX conjugated with branched chain fatty alcohols via disulfide bonds (HUA-SS-DTX) significantly improved the antitumor efficiency of DTX and reduced the systematic toxicity. Our study elaborates on the vital role of connecting bonds and modifying chains in the rational design of prodrug nanoassemblies.
Collapse
Affiliation(s)
- Danping Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chaoying Du
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuo Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingxiao Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiaxuan Song
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinglei Zhai
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bingjun Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
77
|
Ahmad A, Imran M, Sharma N. Precision Nanotoxicology in Drug Development: Current Trends and Challenges in Safety and Toxicity Implications of Customized Multifunctional Nanocarriers for Drug-Delivery Applications. Pharmaceutics 2022; 14:2463. [PMID: 36432653 PMCID: PMC9697541 DOI: 10.3390/pharmaceutics14112463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/06/2022] [Accepted: 11/13/2022] [Indexed: 11/17/2022] Open
Abstract
The dire need for the assessment of human and environmental endangerments of nanoparticulate material has motivated the formulation of novel scientific tools and techniques to detect, quantify, and characterize these nanomaterials. Several of these paradigms possess enormous possibilities for applications in many of the realms of nanotoxicology. Furthermore, in a large number of cases, the limited capabilities to assess the environmental and human toxicological outcomes of customized and tailored multifunctional nanoparticles used for drug delivery have hindered their full exploitation in preclinical and clinical settings. With the ever-compounded availability of nanoparticulate materials in commercialized settings, an ever-arising popular debate has been egressing on whether the social, human, and environmental costs associated with the risks of nanomaterials outweigh their profits. Here we briefly review the various health, pharmaceutical, and regulatory aspects of nanotoxicology of engineered multifunctional nanoparticles in vitro and in vivo. Several aspects and issues encountered during the safety and toxicity assessments of these drug-delivery nanocarriers have also been summarized. Furthermore, recent trends implicated in the nanotoxicological evaluations of nanoparticulate matter in vitro and in vivo have also been discussed. Due to the absence of robust and rigid regulatory guidelines, researchers currently frequently encounter a larger number of challenges in the toxicology assessment of nanocarriers, which have also been briefly discussed here. Nanotoxicology has an appreciable and significant part in the clinical translational development as well as commercialization potential of nanocarriers; hence these aspects have also been touched upon. Finally, a brief overview has been provided regarding some of the nanocarrier-based medicines that are currently undergoing clinical trials, and some of those which have recently been commercialized and are available for patients. It is expected that this review will instigate an appreciable interest in the research community working in the arena of pharmaceutical drug development and nanoformulation-based drug delivery.
Collapse
Affiliation(s)
- Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Mohammad Imran
- Therapeutics Research Group, Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane 4102, Australia
| | - Nisha Sharma
- Division of Nephrology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
78
|
Zhang Y, Shi J, Liu L, Su X, Peng B, Sun W, Li J, Feng Y, Geng Y, Cheng G. Improving Solubility and Avoiding Hygroscopicity of Tetrahydropalmatine by Forming a Pharmaceutical Salt Cocrystal via CAHBs. CRYSTAL RESEARCH AND TECHNOLOGY 2022. [DOI: 10.1002/crat.202200151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Yunan Zhang
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Jingwen Shi
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Lixin Liu
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Xin Su
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Bihui Peng
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Weitong Sun
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Jinjing Li
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Yanru Feng
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | - Yiding Geng
- College of Pharmacy Jiamusi University Jiamusi 154007 China
| | | |
Collapse
|
79
|
Liang S, Wang C, Shao Y, Wang Y, Xing D, Geng Z. Recent advances in bacteria-mediated cancer therapy. Front Bioeng Biotechnol 2022; 10:1026248. [PMID: 36312554 PMCID: PMC9597243 DOI: 10.3389/fbioe.2022.1026248] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Cancer is among the leading cause of deaths worldwide. Although conventional therapies have been applied in the fight against the cancer, the poor oxygen, low extracellular pH, and high interstitial fluid pressure of the tumor microenvironment mean that these treatments fail to completely eradicate cancer cells. Recently, bacteria have increasingly been considered to be a promising platform for cancer therapy thanks to their many unique properties, such as specific tumor-targeting ability, high motility, immunogenicity, and their use as gene or drug carriers. Several types of bacteria have already been used for solid and metastatic tumor therapies, with promising results. With the development of synthetic biology, engineered bacteria have been endowed with the controllable expression of therapeutic proteins. Meanwhile, nanomaterials have been widely used to modify bacteria for targeted drug delivery, photothermal therapy, magnetothermal therapy, and photodynamic therapy, while promoting the antitumor efficiency of synergistic cancer therapies. This review will provide a brief introduction to the foundation of bacterial biotherapy. We begin by summarizing the recent advances in the use of many different types of bacteria in multiple targeted tumor therapies. We will then discuss the future prospects of bacteria-mediated cancer therapies.
Collapse
Affiliation(s)
- Shuya Liang
- Department of Dermatology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Wang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yingchun Shao
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yanhong Wang
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Yanhong Wang, ; Dongming Xing, ; Zhongmin Geng,
| | - Dongming Xing
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
- *Correspondence: Yanhong Wang, ; Dongming Xing, ; Zhongmin Geng,
| | - Zhongmin Geng
- Qingdao Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- *Correspondence: Yanhong Wang, ; Dongming Xing, ; Zhongmin Geng,
| |
Collapse
|
80
|
Endocytosis-mediated triple-activable prodrug nanotherapeutics potentiating therapeutic efficacy and security towards solid tumors. Colloids Surf B Biointerfaces 2022; 218:112723. [DOI: 10.1016/j.colsurfb.2022.112723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 07/12/2022] [Accepted: 07/23/2022] [Indexed: 12/24/2022]
|
81
|
Xing J, Chen W, Chen K, Zhu S, Lin F, Qi Y, Zhang Y, Han S, Rao T, Ruan Y, Zhao S, Yu W, Cheng F. TFAP2C Knockdown Sensitizes Bladder Cancer Cells to Cisplatin Treatment via Regulation of EGFR and NF-κB. Cancers (Basel) 2022; 14:cancers14194809. [PMID: 36230734 PMCID: PMC9562889 DOI: 10.3390/cancers14194809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Bladder cancer (BCa) is considered one of the most common neoplasms of the urology system. Cisplatin-based chemotherapy has been the primary treatment for patients with advanced or metastatic BCa. Nevertheless, cisplatin resistance often limits the treatment of bladder cancer. We expect to find approaches to improve the therapeutic efficacy of cisplatin in bladder cancer. In recent years, many studies have shown that transcription factor AP-2 gamma (TFAP2C) acts as a key player in cancer development and and its expression level is closely related to the sensitivity of tumors to cisplatin. Our study investigated whether TFAP2C affects the sensitivity of BCa cells to cisplatin and the possible mechanisms. We found that TFAP2C expression was significantly upregulated in most BCa tissues compared to adjacent normal tissues. The present study confirmed that TFAP2C knockdown enhanced the anti-tumor effects of cisplatin by decreasing cisplatin-induced activation levels of epidermal growth factor receptor (EGFR) and nuclear factor kappaB (NF-κB). Specifically, this study provides a novel approach to improve the efficacy of cisplatin. Abstract Cisplatin is the first-line chemotherapy for advanced or metastatic bladder cancer. Nevertheless, approximately half of patients with BCa are insensitive to cisplatin therapy or develop cisplatin resistance during the treatment process. Therefore, it is especially crucial to investigate ways to enhance the sensitivity of tumor cells to cisplatin. Transcription factor AP-2 gamma (TFAP2C) is involved in cancer development and chemotherapy sensitivity. However, its relationship with chemotherapy has not been studied in BCa. In this study, we aimed to investigate the therapeutic potential of TFAP2C in human BCa. Results based on TCGA (The Cancer Genome Atlas), GTEx (The Genotype-Tissue Expression) and GEO (Gene Expression Omnibus) data showed that TFAP2C expression was upregulated in BCa tissues and that its high expression was associated with poor prognosis. Meanwhile, we demonstrated the overexpression of TFAP2C in BCa clinical specimens. Subsequently, in vitro, we knocked down TFAP2C in BCa cells and found that TFAP2C knockdown further increased cell cycle arrest and apoptosis caused by cisplatin. In addition, the inhibitory effect of cisplatin on BCa cell migration and invasion was enhanced by TFAP2C knockdown. Our data indicated that cisplatin increased epidermal growth factor receptor (EGFR) and nuclear factor-kappaB (NF-κB) activation levels, but TFAP2C knockdown suppressed this effect. Finally, in vivo data further validated these findings. Our study showed that TFAP2C knockdown affected the activation levels of EGFR and NF-κB and enhanced the anti-tumor effects of cisplatin in vivo and in vitro. This provides a new direction to improve the efficacy of traditional cisplatin chemotherapy.
Collapse
Affiliation(s)
- Ji Xing
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wu Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Kang Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shaoming Zhu
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yucheng Qi
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yunlong Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shangting Han
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Rao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuan Ruan
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Zhao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weimin Yu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (W.Y.); (F.C.)
| | - Fan Cheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Correspondence: (W.Y.); (F.C.)
| |
Collapse
|
82
|
Li B, Zhang X, Li J. Carrier-free supramolecular nanoassemblies of pure LSD1 inhibitor for effective anti-tumor therapy. Front Chem 2022; 10:1012882. [PMID: 36247676 PMCID: PMC9561089 DOI: 10.3389/fchem.2022.1012882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/14/2022] [Indexed: 12/03/2022] Open
Abstract
The LSD1 protein is an oxidase that regulates protein methylation, which regulates gene expression and triggers tumors. Previously, inhibiting LSD1 has been found to be an effective treatment strategy for opposing tumors caused by overexpression of LSD1. Our recent study found that compound 17i was a suitable LSD1 inhibitor with potential anti-tumor activity. However, its extremely insoluble nature limits further validation of its anti-tumor activity at the clinical level. In this study, a unique carrier-free supramolecular nanoassemblies of pure compound 17i is expected to enhance therapeutic efficacy. Aqueous-insoluble compound 17i was mixed with a small quantity of DSPE-PEG2000 into an organic solvent and was prepared as nanoassemblies in water via the one-step nanoprecipitation method. The 17i nanoassemblies have a similar effect on its cytotoxicity when compared with 17i solution in vitro. Importantly, the PEGylated 17i nanoassemblies exhibit significant superiorities over 17i solutions in therapeutic efficiency, anti-tumor immune response and systemic toxicity in BALB/c mice bearing CT-26 colorectal tumors. We envision that the fabrication of pure drug nanoassemblies offers an efficient platform for reforming the undesirable characteristics of drug-like compounds to potentiate the anti-tumor therapeutic effect.
Collapse
Affiliation(s)
- Boao Li
- Department of Colorectal Surgery, Liaoning Cancer Hospital, Shenyang, China
| | - Xiangyu Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- *Correspondence: Xiangyu Zhang, ; Jibin Li,
| | - Jibin Li
- Department of Colorectal Surgery, Liaoning Cancer Hospital, Shenyang, China
- *Correspondence: Xiangyu Zhang, ; Jibin Li,
| |
Collapse
|
83
|
Ding C, Chen C, Zeng X, Chen H, Zhao Y. Emerging Strategies in Stimuli-Responsive Prodrug Nanosystems for Cancer Therapy. ACS NANO 2022; 16:13513-13553. [PMID: 36048467 DOI: 10.1021/acsnano.2c05379] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Prodrugs are chemically modified drug molecules that are inactive before administration. After administration, they are converted in situ to parent drugs and induce the mechanism of action. The development of prodrugs has upgraded conventional drug treatments in terms of bioavailability, targeting, and reduced side effects. Especially in cancer therapy, the application of prodrugs has achieved substantial therapeutic effects. From serendipitous discovery in the early stage to functional design with pertinence nowadays, the importance of prodrugs in drug design is self-evident. At present, studying stimuli-responsive activation mechanisms, regulating the stimuli intensity in vivo, and designing nanoscale prodrug formulations are the major strategies to promote the development of prodrugs. In this review, we provide an outlook of recent cutting-edge studies on stimuli-responsive prodrug nanosystems from these three aspects. We also discuss prospects and challenges in the future development of such prodrugs.
Collapse
Affiliation(s)
- Chendi Ding
- Clinical Research Center, Maoming People's Hospital, 101 Weimin Road, Maoming 525000, China
- School of Medicine, Jinan University, 855 Xingye East Road, Guangzhou 510632, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Chunbo Chen
- Clinical Research Center, Maoming People's Hospital, 101 Weimin Road, Maoming 525000, China
| | - Xiaowei Zeng
- Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Hongzhong Chen
- Institute of Pharmaceutics, School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
84
|
Wang Q, Wang Z, Sun X, Jiang Q, Sun B, He Z, Zhang S, Luo C, Sun J. Lymph node-targeting nanovaccines for cancer immunotherapy. J Control Release 2022; 351:102-122. [PMID: 36115556 DOI: 10.1016/j.jconrel.2022.09.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/20/2022]
Abstract
Cancer immunotherapies such as tumor vaccines, chimeric antigen receptor T cells and immune checkpoint blockades, have attracted tremendous attention. Among them, tumor vaccines prime immune response by delivering antigens and adjuvants to the antigen presenting cells (APCs), thus enhancing antitumor immunotherapy. Despite tumor vaccines have made considerable achievements in tumor immunotherapy, it remains challenging to efficiently deliver tumor vaccines to activate the dendritic cells (DCs) in lymph nodes (LNs). Rational design of nanovaccines on the basis of biomedical nanotechnology has emerged as one of the most promising strategies for boosting the outcomes of cancer immunotherapy. In recent years, great efforts have been made in exploiting various nanocarrier-based LNs-targeting tumor nanovaccines. In view of the rapid advances in this field, we here aim to summarize the latest progression in LNs-targeting nanovaccines for cancer immunotherapy, with special attention to various nano-vehicles developed for LNs-targeting delivery of tumor vaccines, including lipid-based nanoparticles, polymeric nanocarriers, inorganic nanocarriers and biomimetic nanosystems. Moreover, the recent trends in nanovaccines-based combination cancer immunotherapy are provided. Finally, the rationality, advantages and challenges of LNs-targeting nanovaccines for clinical translation and application are spotlighted.
Collapse
Affiliation(s)
- Qiu Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhe Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinxin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Qikun Jiang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shenwu Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
85
|
Shi Z, Liu J, Tian L, Li J, Gao Y, Xing Y, Yan W, Hua C, Xie X, Liu C, Liang C. Insights into stimuli-responsive diselenide bonds utilized in drug delivery systems for cancer therapy. Biomed Pharmacother 2022; 155:113707. [PMID: 36122520 DOI: 10.1016/j.biopha.2022.113707] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Due to the complexity and particularity of cancer cell microenvironments, redox responsive drug delivery systems (DDSs) for cancer therapy have been extensively explored. Compared with widely reported cancer treatment systems based on disulfide bonds, diselenide bonds have better redox properties and greater anticancer efficiency. In this review, the significance and application of diselenide bonds in DDSs are summarized, and the stimulation sensitivity of diselenide bonds is comprehensively reported. The potential and prospects for the application of diselenide bonds in next-generation anticancer drug treatment systems are extensively discussed.
Collapse
Affiliation(s)
- Zhenfeng Shi
- Department of Urology Surgery Center, The People's Hospital of Xinjiang Uyghur Autonomous Region, Urumqi 830002, PR China.
| | - Jifang Liu
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China; College of Life Science, Northwest University, Xi'an 710069, PR China.
| | - Lei Tian
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China; College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Jingyi Li
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Yue Gao
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Yue Xing
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Wenjing Yan
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Chenyu Hua
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| | - Xiaolin Xie
- Shaanxi Panlong Pharmaceutical Group Co., Ltd. Xi'an 710025, PR China.
| | - Chang Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Zhuhai 519030, PR China.
| | - Chengyuan Liang
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi'an 710021, PR China.
| |
Collapse
|
86
|
Dong S, Zhang Y, Guo X, Zhang C, Wang Z, Yu J, Liu Y, Li C, Hu Y, Sun B, Sun M, Zhang H, Ouyang D, He Z, Wang Y. Glutathione Pulse Therapy: Promote Spatiotemporal Delivery of Reduction-Sensitive Nanoparticles at the "Cellular Level" and Synergize PD-1 Blockade Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202744. [PMID: 35896947 PMCID: PMC9507359 DOI: 10.1002/advs.202202744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Spatiotemporal delivery of nanoparticles (NPs) at the "cellular level" is critical for nanomedicine, which is expected to deliver as much cytotoxic drug into cancer cells as possible when NPs accumulate in tumors. However, macrophages and cancer-associated fibroblasts (CAFs) that are present within tumors limit the efficiency of spatiotemporal delivery. To overcome this limitation, glutathion pulse therapy is designed to promote reduction-sensitive Larotaxel (LTX) prodrug NPs to escape the phagocytosis of macrophages and penetrate through the stromal barrier established by CAFs in the murine triple negative breast cancer model. This therapy improves the penetration of NPs in tumor tissues as well as the accumulation of LTX in cancer cells, and remodels the immunosuppressive microenvironment to synergize PD-1 blockade therapy. More importantly, a method is established that can directly observe the biodistribution of NPs between different cells in vivo to accurately quantify the target drugs accumulated in these cells, thereby advancing the spatiotemporal delivery research of NPs at the "cellular level."
Collapse
Affiliation(s)
- Songtao Dong
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Yuan Zhang
- School of PharmacyChina Medical UniversityShenyang110122China
| | - Xiangnan Guo
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Chuang Zhang
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Zhaomeng Wang
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Jiang Yu
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Yubo Liu
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Chang Li
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Yuting Hu
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Bingjun Sun
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Mengchi Sun
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Haotian Zhang
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
| | - Zhonggui He
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| | - Yongjun Wang
- Department of PharmaceuticsWuya College of InnovationShenyang Pharmaceutical UniversityShenyang110016China
| |
Collapse
|
87
|
Ma Z, Lin K, Tang M, Ramachandran M, Qiu R, Li J, Solano LN, Huang Y, De Souza C, Abou-Adas S, Xiang B, Zhang L, Li M, Li Y. A pH-Driven Small-Molecule Nanotransformer Hijacks Lysosomes and Overcomes Autophagy-Induced Resistance in Cancer. Angew Chem Int Ed Engl 2022; 61:e202204567. [PMID: 35791769 PMCID: PMC9995155 DOI: 10.1002/anie.202204567] [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: 03/28/2022] [Indexed: 11/11/2022]
Abstract
Smart conversion of supramolecular structures in vivo is an attractive strategy in cancer nanomedicine, which is usually achieved via specific peptide sequences. Here we developed a lysosomal targeting small-molecule conjugate, PBC, which self-assembles into nanoparticles at physiological pH and smartly converts to nanofibrils in lysosomes of tumor cells. Such a transformation mechanically leads to lysosomal dysfunction, autophagy inhibition, and unusual cytoplasmic vacuolation, thus granting PBC a unique anticancer activity as a monotherapy. Importantly, the photo-activated PBC elicits significant phototoxicity to lysosomes and shows enormous advantages in overcoming autophagy-caused treatment resistance frequently occurring in conventional phototherapy. This improved phototherapy achieves a complete cure of oral cancer xenografts upon limited administration. Our work provides a new paradigm for the construction of nonpeptide nanotransformers with biomedical activities.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University, Jinan, Shandong, 250012, China
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Kai Lin
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, 266003, China
| | - Menghuan Tang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Mythili Ramachandran
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Reng Qiu
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Jin Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Lucas N Solano
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Cristabelle De Souza
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Sara Abou-Adas
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Bai Xiang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| | - Lanwei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, 266003, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine Shandong University, Jinan, Shandong, 250012, China
| | - Yuanpei Li
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA 95817, USA
| |
Collapse
|
88
|
Liu W, Li S, Wang B, Peng P, Gao C. Physiologically Responsive Polyurethanes for Tissue Repair and Regeneration. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Wenxing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Shifen Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Beiduo Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Pai Peng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| |
Collapse
|
89
|
Xiang X, Feng X, Lu S, Jiang B, Hao D, Pei Q, Xie Z, Jing X. Indocyanine green potentiated paclitaxel nanoprodrugs for imaging and chemotherapy. EXPLORATION (BEIJING, CHINA) 2022; 2:20220008. [PMID: 37325605 PMCID: PMC10190853 DOI: 10.1002/exp.20220008] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/12/2022] [Indexed: 06/17/2023]
Abstract
Self-assembled prodrug nanoparticles with tumor-responsive capacity have great potential in tumor visualization and treatment. However, the nanoparticle formulas usually contain multiple components, especially polymeric materials, which result in various potential issues. Herein, we report an indocyanine green (ICG)-driven assembly of paclitaxel prodrugs integrating near-infrared fluorescence imaging and tumor-specific chemotherapy. By feat of the hydrophilic merit of ICG, paclitaxel dimer could form more uniformly monodispersed nanoparticles. This two-in-one strategy reinforces the complementary advantages, resulting in superior assembly behavior, robust colloidal stability, enhanced tumor accumulation as well as desirable near-infrared imaging and in vivo feedback of chemotherapy. The in vivo experiments validated the prodrug activation at tumor sites as evidenced by enhanced fluorescence intensity, potent tumor growth suppression, and reduced systemic toxicity compared with commercial Taxol. The universality of ICG potentiated strategy toward photosensitizers and fluorescence dyes was confirmed. This presentation provides deep insight into the feasibility of constructing clinical-close alternatives for improving antitumor efficacy.
Collapse
Affiliation(s)
- Xiujuan Xiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
- University of Science and Technology of ChinaHefeiChina
| | - Xuan Feng
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
- University of Science and Technology of ChinaHefeiChina
| | - Shaojin Lu
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
- University of Science and Technology of ChinaHefeiChina
| | - Bowen Jiang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
- University of Science and Technology of ChinaHefeiChina
| | - Dengyuan Hao
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
- University of Science and Technology of ChinaHefeiChina
| | - Qing Pei
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
| | - Zhigang Xie
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
- University of Science and Technology of ChinaHefeiChina
| | - Xiabin Jing
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilinChina
| |
Collapse
|
90
|
Wang X, Li C, Wang Y, Chen H, Zhang X, Luo C, Zhou W, Li L, Teng L, Yu H, Wang J. Smart drug delivery systems for precise cancer therapy. Acta Pharm Sin B 2022; 12:4098-4121. [DOI: 10.1016/j.apsb.2022.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/25/2022] [Accepted: 08/08/2022] [Indexed: 11/28/2022] Open
|
91
|
Zhou L, Wu J, Sun Z, Wang W. Oxidation and Reduction Dual-Responsive Polymeric Prodrug Micelles Co-delivery Precisely Prescribed Paclitaxel and Honokiol for Laryngeal Carcinoma Combination Therapy. Front Pharmacol 2022; 13:934632. [PMID: 35935846 PMCID: PMC9354237 DOI: 10.3389/fphar.2022.934632] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/17/2022] [Indexed: 12/05/2022] Open
Abstract
Laryngeal carcinoma is the most common head and neck malignancy globally, and chemotherapy is still the most common treatment for this type of carcinoma. Monotherapy has become powerless because of the lack of drugs in the anticancer agent library, the difficult process of new drug discovery, and the widespread drug resistance. Combination therapy with two agents, in particular Chinese herbal medicines with chemotherapy drugs, is a potential alternative to chemotherapy alone. However, combination therapy faces difficulties in delivering multiple drugs to tumor tissue in a precise ratio. Here, a cocktail polymeric prodrug micelle (PHPPM) was developed using an oxidation and reduction dual-responsive polymeric paclitaxel (PTX) and polymeric honokiol (HK) prodrugs. Both of them were obtained by covalently conjugating the drug to dextran via diselenium bonds. Following optimization and characterization, the PHPPM with the precise mass ratio of PTX and HK was obtained, enabling ratiometric drug loading, synchronized drug release in response to tumor high-level reactive oxygen species and glutathione environment, long blood circulation, and high tumor accumulation. This co-delivery system can effectively inhibit laryngeal carcinoma growth in vitro and in vivo. Codelivery of chemotherapy agents and Chinese herbal medicine with a precise ratio and controlled release of the two drugs at the tumor site provides an effective approach to clinical therapy for other laryngeal carcinomas.
Collapse
|
92
|
Ou D, Ling N, Wang X, Zou Y, Dong J, Zhang D, Shen Y, Ye Y. Safety Assessment of One Lactiplantibacillus plantarum Isolated from the Traditional Chinese Fermented Vegetables—Jiangshui. Foods 2022; 11:foods11152177. [PMID: 35892762 PMCID: PMC9332144 DOI: 10.3390/foods11152177] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 02/01/2023] Open
Abstract
Lactiplantibacillus plantarum is a kind of extensively utilized probiotic species, which plays a critical role in the prevention of pathogenic bacteria and development of functional probiotics. Our group previously isolated one Lactiplantibacillus from Jiang Shui, a traditional Chinese fermented vegetable, which remarkably inhibited the growth of Aspergillus flavus. Herein, the safety of this isolate was assessed to ensure its application feasibility in food industry. Firstly, the phenotypic analyses including tolerance to low pH and bile salt, aggregation ability, and hemolytic activity detection, indicated the isolate could survive and colonize in the gastrointestinal tract, without hemolysin activity. The susceptibilities of the isolate to eight antibiotics and the absence of most resistance genes were demonstrated by agar disk diffusion and PCR, respectively. Furthermore, no mortality or toxicity was observed in mice by in vivo tests using gross autopsy, hematology, serum biochemistry, and HE-staining. Taken together, this study demonstrated the safety of Lactiplantibacillus plantarum WYH as a probiotic strain in terms of phenotypic analyses, absence of antimicrobial resistance and toxin-related genes, as well as mice toxicity test, while supported the prospect of applying isolate in suppression of fungal growth and mycotoxin biosynthesis.
Collapse
|
93
|
Huang Z, Luo Y, Zhang T, Ding Y, Chen M, Chen J, Liu Q, Huang Y, Zhao C. A Stimuli-Responsive Small-Molecule Metal-Carrying Prochelator: A Novel Prodrug Design Strategy for Metal Complexes. Angew Chem Int Ed Engl 2022; 61:e202203500. [PMID: 35513877 DOI: 10.1002/anie.202203500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Indexed: 12/25/2022]
Abstract
Selective activation of prodrugs is an important approach to reduce the side effects of disease treatment. We report a prodrug design concept for metal complexes, termed "metal-carrying prochelator", which can co-carry a metal ion and chelator within a single small-molecule compound and remain inert until it undergoes a specifically triggered intramolecular chelation to synthesize a bioactive metal complex in situ for targeted therapy. As a proof-of-concept, we designed a H2 O2 -responsive small-molecule prochelator, DPBD, based on the strong chelator diethyldithiocarbamate (DTC) and copper. DPBD can carry Cu2+ (DPBD-Cu) and respond to elevated H2 O2 levels in tumor cells by releasing DTC, which rapidly chelates Cu2+ from DPBD-Cu affording a DTC-copper complex with high cytotoxicity, realizing potent antitumor efficacy with low systemic toxicity. Thus, with its unique intramolecularly triggered activation mechanism, this concept based on a small-molecule metal-carrying prochelator can help in the prodrug design of metal complexes.
Collapse
Affiliation(s)
- Zeqian Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yong Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Tao Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yaqing Ding
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Meixu Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jie Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Qiuxing Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yanjuan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Chunshun Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
94
|
Ma Z, Lin K, Tang M, Ramachandran M, Qiu R, Li J, Solano LN, Huang Y, De Souza C, Abou-Adas S, Xiang B, Zhang L, Li M, Li Y. A pH‐Driven Small‐Molecule Nanotransformer Hijacks Lysosomes and Overcomes Autophagy‐Induced Resistance in Cancer. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Zhao Ma
- Shandong University Cheeloo College of Medicine Medicinal Chemistry CHINA
| | - Kai Lin
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Menghuan Tang
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Mythili Ramachandran
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Reng Qiu
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Jin Li
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Lucas N. Solano
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Yanyu Huang
- University of California Davis Biochemistry and Molecular Medicine UNITED STATES
| | - Cristabelle De Souza
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Sara Abou-Adas
- University of California Davis Biochemistry and Molecular Medicine UNITED STATES
| | - Bai Xiang
- University of California Davis School of Medicine Biochemistry and Molecular Medicine UNITED STATES
| | - Lanwei Zhang
- Ocean University of China College of Food Science and Engineering CHINA
| | - Minyong Li
- Shandong University Cheeloo College of Medicine Medicinal Chemistry CHINA
| | - Yuanpei Li
- University of California Davis School of Medicine Biochemistry and Molecular Medicine 2700 Stockton Blvd, Suite 2405 95817 Sacramento UNITED STATES
| |
Collapse
|
95
|
Long chain triglyceride-lipid formulation promotes the oral absorption of the lipidic prodrugs through coincident intestinal behaviors. Eur J Pharm Biopharm 2022; 176:122-132. [DOI: 10.1016/j.ejpb.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/24/2022] [Accepted: 05/21/2022] [Indexed: 11/22/2022]
|
96
|
Dutta G, Manickam S, Sugumaran A. Stimuli-Responsive Hybrid Metal Nanocomposite - A Promising Technology for Effective Anticancer Therapy. Int J Pharm 2022; 624:121966. [PMID: 35764265 DOI: 10.1016/j.ijpharm.2022.121966] [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: 05/04/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 11/19/2022]
Abstract
Cancer is one of the most challenging, life-threatening illnesses to cure, with over 10 million new cases diagnosed each year globally. Improved diagnostic cum treatment with common side-effects are warranting for successful therapy. Nanomaterials are recognized to improve early diagnosis, imaging, and treatment. Recently, multifunctional nanocomposites attracted considerable interest due to their low-cost production, and ideal thermal and chemical stability, and will be beneficial in future diagnostics and customized treatment capacity. Stimuli-Responsive Hybrid Metal Nanocomposites (SRHMNs) based nanocomposite materials pose the on/off delivery of bioactive compounds such as medications, genes, RNA, and DNA to specific tissue or organs and reduce toxicity. They simultaneously serve as sophisticated imaging and diagnostic tools when certain stimuli (e.g., temperature, pH, redox, ultrasound, or enzymes) activate the nanocomposite, resulting in the imaging-guided transport of the payload at defined sites. This review in detail addresses the recent advancements in the design and mechanism of internal breakdown processes of the functional moiety from stimuli-responsive systems in response to a range of stimuli coupled with metal nanoparticles. Also, it provides a thorough understanding of SRHMNs, enabling non-invasive interventional therapy by resolving several difficulties in cancer theranostics.
Collapse
Affiliation(s)
- Gouranga Dutta
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Sivakumar Manickam
- Petroleum and Chemical Engineering, Faculty of Engineering, Universiti Teknologi Brunei, Jalan Tungku Link Gadong, BE1410, Brunei Darussalam
| | - Abimanyu Sugumaran
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, India.
| |
Collapse
|
97
|
Sun L, Zhao P, Chen M, Leng J, Luan Y, Du B, Yang J, Yang Y, Rong R. Taxanes prodrug-based nanomedicines for cancer therapy. J Control Release 2022; 348:672-691. [PMID: 35691501 DOI: 10.1016/j.jconrel.2022.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/04/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022]
Abstract
Malignant tumor remains a huge threat to human health and chemotherapy still occupies an important place in clinical tumor treatment. As a kind of potent antimitotic agent, taxanes act as the first-line broad-spectrum cancer drug in clinical use. However, disadvantages such as prominent hydrophobicity, severe off-target toxicity or multidrug resistance lead to unsatisfactory therapeutic effects, which restricts its wider usage. The efficient delivery of taxanes is still quite a challenge despite the rapid developments in biomaterials and nanotechnology. Great progress has been made in prodrug-based nanomedicines (PNS) for cancer therapy due to their outstanding advantages such as high drug loading efficiency, low carrier induced immunogenicity, tumor stimuli-responsive drug release, combinational therapy and so on. Based on the numerous developments in this filed, this review summarized latest updates of taxanes prodrugs-based nanomedicines (TPNS), focusing on polymer-drug conjugate-based nanoformulations, small molecular prodrug-based self-assembled nanoparticles and prodrug-encapsulated nanosystems. In addition, the new trends of tumor stimuli-responsive TPNS were also discussed. Moreover, the future challenges of TPNS for clinical translation were highlighted. We here expect this review will inspire researchers to explore more practical taxanes prodrug-based nano-delivery systems for clinical use.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Menghan Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jiayi Leng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yixin Luan
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jia Yang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
98
|
Zhao R, Ning X, Wang M, Wang H, Xing G, Wang L, Lu C, Yu A, Wang Y. A ROS-Responsive Simvastatin Nano-Prodrug and its Fibronectin-Targeted Co-Delivery System for Atherosclerosis Treatment. ACS APPLIED MATERIALS & INTERFACES 2022; 14:25080-25092. [PMID: 35618653 DOI: 10.1021/acsami.2c02354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nanoprodrugs with responsive release properties integrate the advantages of stimuli-responsive prodrugs and nanotechnology. They would provide ultimate opportunity in fighting atherosclerosis. In this study, we synthesized a redox-responsive nanoprodrug of simvastatin (TPTS) by conjugating α-tocopherol polyethylene glycol derivative to the pharmacophore of simvastatin with a thioketal linker. TPTS formed nanoparticles and released parent simvastatin in the presence of hydrogen peroxide. Moreover, by taking advantage of the self-assembly behavior of TPTS, we developed a fibronectin-targeted delivery system (TPTS/C/T) to codelivery simvastatin prodrug and ticagrelor. In vitro and in vivo experiments indicated that TPTS and TPTS/C/T had good stability, which could reduce off-target leakage of drugs. They greatly inhibited the M1-type polarization of macrophages; reduced intracellular reactive oxygen species level and inflammatory cytokine; and TNF-α, MCP-1, and IL-1β were secreted by macrophage cells, thus providing enhanced anti-inflammatory and antioxidant effects compared with free simvastatin. TPTS/C/T realized targeted drug release to plaques and synergistic therapeutic effects of simvastatin and ticagrelor on atherosclerosis treatment in an ApoE-/- mouse model, resulting in excellent atherosclerosis therapeutic efficacy and a promising biosafety profile. Therefore, this study provides a new method for manufacturing statin nanodrugs and a new design idea for related responsive drug release nanosystems for atherosclerosis.
Collapse
Affiliation(s)
- Runze Zhao
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xiaoyue Ning
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Mengqi Wang
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Huanhuan Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Guang Xing
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Li Wang
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Chengzhi Lu
- Department of Cardiology, Tianjin First Central Hospital, Tianjin 300192, China
| | - Ao Yu
- College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yongjian Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
99
|
Ren G, Duan D, Wang G, Wang R, Li Y, Zuo H, Zhang Q, Zhang G, Zhao Y, Wang R, Zhang S. Construction of reduction-sensitive heterodimer prodrugs of doxorubicin and dihydroartemisinin self-assembled nanoparticles with antitumor activity. Colloids Surf B Biointerfaces 2022; 217:112614. [PMID: 35700564 DOI: 10.1016/j.colsurfb.2022.112614] [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: 03/02/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 11/20/2022]
Abstract
Doxorubicin (DOX) is used as a first-line chemotherapeutic drug, whereas dihydroartemisinin (DHA) also shows a certain degree of antitumor activity. Disulfide bonds (-SS-) in prodrug molecules can be degraded in highly reducing environments. Thus, heterodimer prodrugs of DOX and DHA linked by a disulfide bond was designed and subsequently prepared as reduction-responsive self-assembled nanoparticles (DOX-SS-DHA NPs). In an in vitro release study, DOX-SS-DHA NPs exhibited reduction-responsive activity. Upon cellular evaluation, DOX-SS-DHA NPs were found to have better selectivity toward tumor cells and less cytotoxicity to normal cells. Compared to free DiR, DOX-SS-DHA NPs showed improved accumulation at the tumor site and even had a longer clearance half-life. More importantly, DOX-SS-DHA NPs possessed a much higher tumor inhibition efficacy than DOX-sol and MIX-sol in 4T1 tumor-bearing mice. Our results suggested the superior antitumor efficacy of DOX-SS-DHA NPs with less cytotoxicity.
Collapse
Affiliation(s)
- Guolian Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Danyu Duan
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Geng Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rongrong Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yujie Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hengtong Zuo
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qichao Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guoshun Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongdan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruili Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
100
|
Xie YJ, Huang M, Li D, Hou JC, Liang HH, Nasim AA, Huang JM, Xie C, Leung ELH, Fan XX. Bacteria-based nanodrug for anticancer therapy. Pharmacol Res 2022; 182:106282. [PMID: 35662630 DOI: 10.1016/j.phrs.2022.106282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 12/15/2022]
Abstract
Bacteria-based immunotherapy has become a promising strategy to induce innate and adaptive responses for fighting cancer. The advantages of bacteriolytic tumor therapy mainly lie in stimulation of innate immunity and colonization of some bacteria targeting the tumor microenvironment (TME). These bacteria have cytotoxic proteins and immune modulating factors that can effectively restrain tumor growth. However, cancer is a multifactorial disease and single therapy is typically unable to eradicate tumors. Rapid progress has been made in combining bacteria with nanotechnology. Using the nanomolecular properties of bacterial products for tumor treatment preserves many features from the original bacteria while providing some unique advantages. Nano-bacterial therapy can enhance permeability and retention of drugs, increase the tolerability of the targeted drugs, promote the release of immune cell mediators, and induce immunogenic cell death pathways. In addition, combining nano-bacterial mediated antitumor therapeutic systems with modern therapy is an effective strategy for overcoming existing barriers in antitumor treatment and can achieve satisfactory therapeutic efficacy. Overall, exploring the immune antitumor characteristics of adjuvant clinical treatment with bacteria can provide potential efficacious treatment strategies for combatting cancer.
Collapse
Affiliation(s)
- Ya-Jia Xie
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Min Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Dan Li
- Beijing Wante'er Biological Pharmaceutical Co., Ltd., No. 32 Yard, East 2nd Road, Yanqi Economic Development Zone, Huairou District, Beijing, China
| | - Jin-Cai Hou
- Beijing Wante'er Biological Pharmaceutical Co., Ltd., No. 32 Yard, East 2nd Road, Yanqi Economic Development Zone, Huairou District, Beijing, China
| | - Hai-Hai Liang
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, China
| | - Ali Adnan Nasim
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Ju-Min Huang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Chun Xie
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Elaine Lai-Han Leung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Xing-Xing Fan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| |
Collapse
|