1
|
Liu J, Tang W, Chen L, Zhang Q, Liu T, Qin L, Zhang Y, Chen X. Engineered gold nanoparticles for accurate and full-scale tumor treatment via pH-dependent sequential charge-reversal and copper triggered photothermal-chemodynamic-immunotherapy. Biomaterials 2025; 321:123322. [PMID: 40222257 DOI: 10.1016/j.biomaterials.2025.123322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/12/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
Current anti-tumor strategies majorly rely on the targeted delivery of functional nanomedicines to tumor region, neglecting the importance of effective infiltration of these nanomedicines in whole tumor tissue. This process normally causes the quick endocytosis by the tumor cells at surface layer of tumor tissue, resulting in the restriction of the penetration of these nanomedicines and limited therapeutic region, which would not be able to treat the entire tumor tissue. Herein, we prepared a series of engineered gold nanoparticles (Au-MBP NPs) with step-wise charge reversal in different acid environments that could entirely infiltrate into the whole tumor tissue and then perform tumor-specific photothermal-chemodynamic-immunotherapy to achieve the complete and accurate tumor treatment. These Au-MBP NPs consisted of AuNPs, thiol modified piperidine (SH-PD, charge reversal group), thiol modified benzoyl thiourea (SH-BTU, copper chelator) and 11-mercaptoundecanoic acid (MUA) with different proportions. Once these Au-MBP NPs arrived tumor tissue, the decreasing pH values from shallow to deep region of tumor tissue separately induced the charge reversal of these nanoparticles from negative to positive, allowing them to bind with negatively charged tumor cells at designed area to occupy the whole tumor for further therapy. Following with the internalization by tumor cells, these Au-MBP NPs would selectively capture the excessive Cu2+ to decrease the available copper in tumor cells, resulting in the inhibition of tumor metastasis via the copper metabolism blockade. On one hand, the captured Cu2+ also induced the aggregation of Au-MBP NPs, which in situ generated the photothermal agents in tumor cells for tumor-specific photothermal therapy (PTT). On the other hand, the chelated Cu2+ ions were reduced to Cu+, which catalyzed the high concentration of intracellular H2O2 to produce cytotoxic hydroxyl radical (•OH), exerting tumor-specific chemodynamic therapy (CDT). Furthermore, the immune-associated tumor antigens were also generated during PTT and CDT processes via immunogenic cell death (ICD), which further matured the dendritic cells (DCs) and then activated CD4+ and CD8+ T cells to turn on the immunotherapy, resulting in additional anti-tumor and anti-metastasis effects. Both in vitro and in vivo results indicated that these Au-MBP NPs possessed enormous potential for effectively suppressing primary and metastatic tumors.
Collapse
Affiliation(s)
- Jie Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wenjuan Tang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qianqian Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tao Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Longyu Qin
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
2
|
Lin Y, Lin P, Xu R, Chen X, Lu Y, Zheng J, Zheng Y, Zhou Z, Mai Z, Zhao X, Cui L. Nanovaccines empowering CD8 + T cells: a precision strategy to enhance cancer immunotherapy. Theranostics 2025; 15:3098-3121. [PMID: 40083941 PMCID: PMC11898294 DOI: 10.7150/thno.107856] [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: 11/29/2024] [Accepted: 01/20/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer immunotherapy leveraging nanovaccines represents a cutting-edge frontier in precision medicine, specifically designed to potentiate CD8+ T cell-based immunotherapy. This review thoroughly delineates the evolving landscape of cancer nanovaccine development, emphasizing their advantageous role in modulating the immunosuppressive tumor microenvironment (TME) to enhance CD8+ T cell efficacy. We critically analyze current innovations in nanovaccine design, focusing on their capacity to deliver tumor antigens and immunostimulatory adjuvants effectively. These nanovaccines are engineered to overcome the physical and immunological barriers of the TME, facilitating the robust activation and proliferation of CD8+ T cells. Challenges such as delivery efficacy, safety, and scalable manufacturing are discussed, alongside future prospects which include the potential of developing specific biomaterial approaches to provide durable antitumor immunity. This comprehensive analysis not only underscores the transformative potential of cancer nanovaccines in enhancing CD8+ T cell responses but also highlights the critical need for advanced solutions to overcome the complex interplay of factors that limit the efficacy of current immunotherapies.
Collapse
Affiliation(s)
- Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Rongwei Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Zihao Zhou
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
- School of Dentistry, University of California, Los Angeles, Los Angeles, 90095, CA, USA
| |
Collapse
|
3
|
Xiong G, Chen Q, Wang Q, Wang X, Xiao Y, Jin L, Yan K, Zhang X, Hu F. Multifaceted role of nanocomposite hydrogels in diabetic wound healing: enhanced biomedical applications and detailed molecular mechanisms. Biomater Sci 2024; 12:6196-6223. [PMID: 39494707 DOI: 10.1039/d4bm01088d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
The complex microenvironment of diabetic wounds, which is characterized by persistent hyperglycemia, excessive inflammatory responses, and hypoxic conditions, significantly impedes the efficacy of traditional hydrogels. Nanocomposite hydrogels, which combine the high-water content and biocompatibility of hydrogels with the unique functionalities of nanomaterials, offer a promising solution. These hydrogels exhibit enhanced antibacterial, antioxidant, and drug-release properties. Incorporating nanomaterials increases the mechanical strength and bioactivity of hydrogels, allowing for dynamic regulation of the wound microenvironment and promoting cell migration, proliferation, and angiogenesis, thereby accelerating wound healing. This review provides a comprehensive overview of the latest advances in nanocomposite hydrogels for diabetic wound treatment and discusses their advantages and molecular mechanisms at various healing stages. The study aims to provide a theoretical foundation and practical guidance for future research and clinical applications. Furthermore, it highlights the challenges related to the mechanical durability, antimicrobial performance, resistance issues, and interactions with the cellular environments of these hydrogels. Future directions include optimizing smart drug delivery systems and personalized medical approaches to enhance the clinical applicability of nanocomposite hydrogels.
Collapse
Affiliation(s)
- Gege Xiong
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Qiwei Chen
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Qiuyu Wang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Xiaoxue Wang
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan 528000, PR China.
| | - Yaomu Xiao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Liuli Jin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Kaichong Yan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| | - Xueyang Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
- Department of Stomatology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde, Foshan), Foshan 528000, PR China.
| | - Fei Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, PR China.
| |
Collapse
|
4
|
Chen J, Yao Y, Mao X, Chen Y, Ni F. Liver-targeted delivery based on prodrug: passive and active approaches. J Drug Target 2024; 32:1155-1168. [PMID: 39072411 DOI: 10.1080/1061186x.2024.2386416] [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: 03/20/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The liver, a central organ in human metabolism, is often the primary target for drugs. However, conditions such as viral hepatitis, cirrhosis, non-alcoholic fatty liver disease (NAFLD), and hepatocellular carcinoma (HCC) present substantial health challenges worldwide. Existing treatments, which suffer from the non-specific distribution of drugs, frequently fail to achieve desired efficacy and safety, risking unnecessary liver harm and systemic side effects. PURPOSE The aim of this review is to synthesise the latest progress in the design of liver-targeted prodrugs, with a focus on passive and active targeting strategies, providing new insights into the development of liver-targeted therapeutic approaches. METHODS This study conducted an extensive literature search through databases like Google Scholar, PubMed, Web of Science, and China National Knowledge Infrastructure (CNKI), systematically collecting and selecting recent research on liver-targeted prodrugs. The focus was on targeting mechanisms, including the Enhanced Permeability and Retention (EPR) effect, the unique microenvironment of liver cancer, and active targeting through specific transporters and receptors. RESULTS Active targeting strategies achieve precise drug delivery by binding specific ligands to liver surface receptors. Passive targeting takes advantage of the EPR effect and tumour characteristics to enrich drugs in liver tumours. The review details successful cases of using small molecule ligands, peptides, antibodies and nanoparticles as drug carriers. CONCLUSION Liver-targeted prodrug strategies show great potential in enhancing the efficacy of drug treatment and reducing side effects for liver diseases. Future research should balance the advantages and limitations of both targeting strategies, focusing on optimising drug design and targeting efficiency, especially for clinical application. In-depth research on liver-specific receptors and the development of innovative targeting molecules are crucial for advancing the field of liver-targeted prodrugs.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingrui Yao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoran Mao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzhou Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| |
Collapse
|
5
|
Swarna MR, Opi MH, Ahmed T, Piya AA, Habiba U, Shamim SUD. Understanding the adsorption performance of hetero-nanocages (C 12-B 6N 6, C 12-Al 6N 6, and B 6N 6-Al 6N 6) towards hydroxyurea anticancer drug: a comprehensive study using DFT. NANOSCALE ADVANCES 2024:d4na00472h. [PMID: 39372438 PMCID: PMC11447748 DOI: 10.1039/d4na00472h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/08/2024] [Indexed: 10/08/2024]
Abstract
Cancer is a paramount health challenge to global health, which forms tumors that can invade nearby tissues and spread to neighboring cells. Recently, nanotechnology has been used to control the growth of cancer, in which anticancer drugs are delivered to cancerous cells via nanoparticles without damaging healthy tissues. In this study, DFT investigations were carried out to examine the adsorption behavior of C24, B12N12, and Al12N12 nanocages as well as their heterostructures C12-B6N6, C12-Al6N6, and B6N6-Al6N6 towards the hydroxyurea (HU) anticancer drug. In this regard, adsorption energy, interaction distance between the drug and nanocages, charge transfer, energy gap, dipole moment, quantum molecular descriptors, work function, and COSMO surface analysis were analyzed to understand their adsorption performance. Findings demonstrate that the adsorption energies of two hetero-nanocages on their hexagonal (SH) and tetragonal (ST) sites are favorable for the drug delivery process. The computed adsorption energy of B6N6-Al6N6 of the ST/AlN site is 183.59 kJ mol-1, which is higher than that of the C12-Al6N6 nanocage, including minimum adsorption distances. Negative adsorption energy with low adsorption distances implies an attractive interaction between the drug and nanocages. During the interaction, a significant amount of charge is transferred between the drug and nanocages. Furthermore, for both complexes, larger dipole moments were observed in water media compared to gas media. From DOS spectra, prominent peaks were found in the Fermi level after adsorption of HU on the nanocages, implying the reduction of the energy gap. Noticeable overlaps between the PDOS spectra of the nanocages and HU's close contact atom demonstrate the formation of chemical bonds between two specific atoms. Therefore, it can be concluded that among the nanocages, C12-Al6N6 and B6N6-Al6N6 may be suitable carriers for HU drug.
Collapse
Affiliation(s)
- Mithila Roy Swarna
- Department of Physics, Mawlana Bhashani Science and Technology University Tangail Bangladesh
| | - Mehedi Hasan Opi
- Department of Physics, Mawlana Bhashani Science and Technology University Tangail Bangladesh
| | - Tanvir Ahmed
- Department of Physics, Mawlana Bhashani Science and Technology University Tangail Bangladesh
| | - Afiya Akter Piya
- Department of Physics, Mawlana Bhashani Science and Technology University Tangail Bangladesh
| | - Umme Habiba
- Department of Physics, Mawlana Bhashani Science and Technology University Tangail Bangladesh
| | - Siraj Ud Daula Shamim
- Department of Physics, Mawlana Bhashani Science and Technology University Tangail Bangladesh
| |
Collapse
|
6
|
Xu M, Feng G, Fang J. Microcapsules based on biological macromolecules for intestinal health: A review. Int J Biol Macromol 2024; 276:133956. [PMID: 39029830 DOI: 10.1016/j.ijbiomac.2024.133956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Intestinal dysfunction is becoming increasingly associated with neurological and endocrine issues, raising concerns about its impact on world health. With the introduction of several breakthrough technologies for detecting and treating intestinal illnesses, significant progress has been made in the previous few years. On the other hand, traditional intrusive diagnostic techniques are expensive and time-consuming. Furthermore, the efficacy of conventional drugs (not capsules) is reduced since they are more likely to degrade before reaching their target. In this context, microcapsules based on different types of biological macromolecules have been used to encapsulate active drugs and sensors to track intestinal ailments and address these issues. Several biomacromolecules/biomaterials (natural protein, alginate, chitosan, cellulose and RNA etc.) are widely used for make microcapsules for intestinal diseases, and can significantly improve the therapeutic effect and reduce adverse reactions. This article systematically summarizes microencapsulated based on biomacromolecules material for intestinal health control and efficacy enhancement. It also discusses the application and mechanism research of microencapsulated biomacromolecules drugs in reducing intestinal inflammation, in addition to covering the preparation techniques of microencapsulated drug delivery systems used for intestinal health. Microcapsule delivery systems' limits and potential applications for intestinal disease diagnosis, treatment, and surveillance were highlighted.
Collapse
Affiliation(s)
- Minhui Xu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha 410128, Hunan, China
| | - Guangfu Feng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha 410128, Hunan, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha 410128, Hunan, China
| |
Collapse
|
7
|
Zhang S, Mou F, Yu Z, Li L, Yang M, Zhang D, Ma H, Luo W, Li T, Guan J. Heterogeneous Sensor-Carrier Microswarms for Collaborative Precise Drug Delivery toward Unknown Targets with Localized Acidosis. NANO LETTERS 2024; 24:5958-5967. [PMID: 38738749 DOI: 10.1021/acs.nanolett.4c00162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Micro/nanorobots hold the potential to revolutionize biomedicine by executing diverse tasks in hard-to-reach biological environments. Nevertheless, achieving precise drug delivery to unknown disease sites using swarming micro/nanorobots remains a significant challenge. Here we develop a heterogeneous swarm comprising sensing microrobots (sensor-bots) and drug-carrying microrobots (carrier-bots) with collaborative tasking capabilities for precise drug delivery toward unknown sites. Leveraging robust interspecific hydrodynamic interactions, the sensor-bots and carrier-bots spontaneously synchronize and self-organize into stable heterogeneous microswarms. Given that the sensor-bots can create real-time pH maps employing pH-responsive structural-color changes and the doxorubicin-loaded carrier-bots exhibit selective adhesion to acidic targets via pH-responsive charge reversal, the sensor-carrier microswarm, when exploring unknown environments, can detect and localize uncharted acidic targets, guide itself to cover the area, and finally deploy therapeutic carrier-bots precisely there. This versatile platform holds promise for treating diseases with localized acidosis and inspires future theranostic microsystems with expandability, task flexibility, and high efficiency.
Collapse
Affiliation(s)
- Shuming Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Fangzhi Mou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Zheng Yu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Luolin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Manyi Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Di Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
| | - Huiru Ma
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
- Wuhan Institute of Photochemistry and Technology, 7 North Bingang Road, Wuhan 430083, People's Republic of China
| | - Wei Luo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
- Wuhan Institute of Photochemistry and Technology, 7 North Bingang Road, Wuhan 430083, People's Republic of China
| | - Tianlong Li
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, People's Republic of China
| | - Jianguo Guan
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, People's Republic of China
- Wuhan Institute of Photochemistry and Technology, 7 North Bingang Road, Wuhan 430083, People's Republic of China
| |
Collapse
|
8
|
Deng H, Zhang Y, Cai X, Yin Z, Yang Y, Dong Q, Qiu Y, Chen Z. Dual-Targeted Graphitic Cascade Nanozymes for Recognition and Treatment of Helicobacter pylori. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306155. [PMID: 37991257 DOI: 10.1002/smll.202306155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/08/2023] [Indexed: 11/23/2023]
Abstract
Helicobacter pylori (H. pylori) is the major etiological factor of a variety of gastric diseases. However, the treatment of H. pylori is challenged by the destruction of targeted drugs by gastric acid and pepsin. Herein, a dual-targeted cascade catalytic nanozyme PtCo@Graphene@Hemin-2(L-arginine) (PtCo@G@H2A) is designed for the treatment of H. pylori. The dual-targeting ability of PtCo@G@H2A is derived from directly targeting the receptor protein of H. pylori through hemin and responding to the acidic environment to cause charge reversal (protonation of L-arginine) to capture H. pylori, achieving efficient targeting effect. Compared with the single-targeting strategy relying on hemin, the dual-targeting strategy can greatly improve the targeting rate, achieving an increase of 850% targeting rate. At the concentration of NaHCO3 in intestinal fluid, the surface potential of PtCo@G@H2A can be quickly restored to avoid side effects. Meanwhile, PtCo@G@H2A has pH-responsive oxidase-like activity, which can generate nitric oxide (NO) through a cascade catalytic process that first generates reactive oxygen species (ROS) with oxygen, and further oxidizes L-arginine through ROS, realizing a superior acid-selective bactericidal effect. Overall, it proposes a promising strategy for the treatment of H. pylori that maintains high targeting and therapeutic effects in the environment of gastric acid and pepsin.
Collapse
Affiliation(s)
- Hui Deng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yi Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Xinqi Cai
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Zhiwei Yin
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Yanxia Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Qian Dong
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Ye Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| | - Zhuo Chen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, 410082, China
| |
Collapse
|
9
|
Luo F, Li M, Chen Y, Song S, Yu H, Zhang P, Xiao C, Lv G, Chen X. Immunosuppressive enzyme-responsive nanoparticles for enhanced accumulation in liver allograft to overcome acute rejection. Biomaterials 2024; 306:122476. [PMID: 38266349 DOI: 10.1016/j.biomaterials.2024.122476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/26/2024]
Abstract
Acute rejection is a life-threatening complication after liver transplantation. Immunosuppressants such as tacrolimus are used to inhibit acute rejection of liver grafts in clinic. However, inefficient intragraft accumulation may reduce the therapeutic outcomes of tacrolimus. Here, an enzyme-responsive nanoparticle is developed to selectively enhance the accumulation of tacrolimus in liver allograft through enzyme-induced aggregation to refine immunotherapeutic efficacy of tacrolimus. The nanoparticles are composed of amphiphilic tacrolimus prodrugs synthesized by covalently conjugating tacrolimus and matrix metalloproteinase 9 (MMP9)-cleavable peptide-containing methoxy poly (ethylene glycol) to poly (l-glutamic acid). Upon exposure to MMP9, which is overexpressed in rejected liver allografts, the nanoparticles undergo a morphological transition from spherical micellar nanoparticles to microscale aggregate-like scaffolds. Intravenous administration of MMP9-responsive nanoparticles into a rat model of acute liver graft rejection results in enhanced nanoparticle accumulation in allograft as compared to nonresponsive nanoparticles. Consequently, the MMP9-responsive nanoparticles significantly inhibit intragraft inflammatory cell infiltration and proliferation, maintain intragraft immunosuppressive environment, alleviate graft damage, improve liver allograft function, abate weight loss and prolong recipient survival. This work proves that morphology-switchable enzyme-responsive nanoparticles represent an innovative strategy for selectively enhancing intragraft accumulation of immunosuppressive agents to improve treatment of liver allograft rejection.
Collapse
Affiliation(s)
- Feixiang Luo
- General Surgery Center, Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Mingqian Li
- General Surgery Center, Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Yuguo Chen
- General Surgery Center, Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Shifei Song
- General Surgery Center, Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| | - Guoyue Lv
- General Surgery Center, Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, 130021, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| |
Collapse
|
10
|
Xu L, Cao Y, Xu Y, Li R, Xu X. Redox-Responsive Polymeric Nanoparticle for Nucleic Acid Delivery and Cancer Therapy: Progress, Opportunities, and Challenges. Macromol Biosci 2024; 24:e2300238. [PMID: 37573033 DOI: 10.1002/mabi.202300238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/25/2023] [Indexed: 08/14/2023]
Abstract
Cancer development and progression of cancer are closely associated with the activation of oncogenes and loss of tumor suppressor genes. Nucleic acid drugs (e.g., siRNA, mRNA, and DNA) are widely used for cancer therapy due to their specific ability to regulate the expression of any cancer-associated genes. However, nucleic acid drugs are negatively charged biomacromolecules that are susceptible to serum nucleases and cannot cross cell membrane. Therefore, specific delivery tools are required to facilitate the intracellular delivery of nucleic acid drugs. In the past few decades, a variety of nanoparticles (NPs) are designed and developed for nucleic acid delivery and cancer therapy. In particular, the polymeric NPs in response to the abnormal redox status in cancer cells have garnered much more attention as their potential in redox-triggered nanostructure dissociation and rapid intracellular release of nucleic acid drugs. In this review, the important genes or signaling pathways regulating the abnormal redox status in cancer cells are briefly introduced and the recent development of redox-responsive NPs for nucleic acid delivery and cancer therapy is systemically summarized. The future development of NPs-mediated nucleic acid delivery and their challenges in clinical translation are also discussed.
Collapse
Affiliation(s)
- Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Ya Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
| | - Rong Li
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| | - Xiaoding Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan, 528200, P. R. China
- The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, P. R. China
| |
Collapse
|
11
|
Shen X, Pan D, Gong Q, Gu Z, Luo K. Enhancing drug penetration in solid tumors via nanomedicine: Evaluation models, strategies and perspectives. Bioact Mater 2024; 32:445-472. [PMID: 37965242 PMCID: PMC10641097 DOI: 10.1016/j.bioactmat.2023.10.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Effective tumor treatment depends on optimizing drug penetration and accumulation in tumor tissue while minimizing systemic toxicity. Nanomedicine has emerged as a key solution that addresses the rapid clearance of free drugs, but achieving deep drug penetration into solid tumors remains elusive. This review discusses various strategies to enhance drug penetration, including manipulation of the tumor microenvironment, exploitation of both external and internal stimuli, pioneering nanocarrier surface engineering, and development of innovative tactics for active tumor penetration. One outstanding strategy is organelle-affinitive transfer, which exploits the unique properties of specific tumor cell organelles and heralds a potentially transformative approach to active transcellular transfer for deep tumor penetration. Rigorous models are essential to evaluate the efficacy of these strategies. The patient-derived xenograft (PDX) model is gaining traction as a bridge between laboratory discovery and clinical application. However, the journey from bench to bedside for nanomedicines is fraught with challenges. Future efforts should prioritize deepening our understanding of nanoparticle-tumor interactions, re-evaluating the EPR effect, and exploring novel nanoparticle transport mechanisms.
Collapse
Affiliation(s)
- Xiaoding Shen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
12
|
Veider F, Sanchez Armengol E, Bernkop-Schnürch A. Charge-Reversible Nanoparticles: Advanced Delivery Systems for Therapy and Diagnosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304713. [PMID: 37675812 DOI: 10.1002/smll.202304713] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/24/2023] [Indexed: 09/08/2023]
Abstract
The past two decades have witnessed a rapid progress in the development of surface charge-reversible nanoparticles (NPs) for drug delivery and diagnosis. These NPs are able to elegantly address the polycation dilemma. Converting their surface charge from negative/neutral to positive at the target site, they can substantially improve delivery of drugs and diagnostic agents. By specific stimuli like a shift in pH and redox potential, enzymes, or exogenous stimuli such as light or heat, charge reversal of NP surface can be achieved at the target site. The activated positive surface charge enhances the adhesion of NPs to target cells and facilitates cellular uptake, endosomal escape, and mitochondrial targeting. Because of these properties, the efficacy of incorporated drugs as well as the sensitivity of diagnostic agents can be essentially enhanced. Furthermore, charge-reversible NPs are shown to overcome the biofilm formed by pathogenic bacteria and to shuttle antibiotics directly to the cell membrane of these microorganisms. In this review, the up-to-date design of charge-reversible NPs and their emerging applications in drug delivery and diagnosis are highlighted.
Collapse
Affiliation(s)
- Florina Veider
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| | - Eva Sanchez Armengol
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| | - Andreas Bernkop-Schnürch
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| |
Collapse
|
13
|
Singh N, Marwaha D, Gautam S, Rai N, Tiwari P, Sharma M, Shukla RP, Mugale MN, Kumar A, Mishra PR. Surface-Modified Lyotropic Crystalline Nanoconstructs Bearing Doxorubicin and Buparvaquone Target Sigma Receptors through pH-Sensitive Charge Conversion to Improve Breast Cancer Therapy. Biomacromolecules 2023; 24:5780-5796. [PMID: 38006339 DOI: 10.1021/acs.biomac.3c00795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
In the current study, we aimed to develop lyotropic crystalline nanoconstructs (LCNs) based on poly(l-glutamic acid) (PLG) with a two-tier strategy. The first objective was to confer pH-responsive charge conversion properties to facilitate the delivery of both doxorubicin (DOX) and buparvaquone (BPQ) in combination (B + D@LCNs) to harness their synergistic effects. The second goal was to achieve targeted delivery to sigma receptors within the tumor tissues. To achieve this, we designed a pH-responsive charge conversion system using a polymer consisting of poly(ethylenimine), poly(l-lysine), and poly(l-glutamic acid) (PLG), which was then covalently coupled with methoxybenzamide (MBA) for potential sigma receptor targeting. The resulting B + D@LCNs were further modified by surface functionalization with PLG-MBA to confer both sigma receptor targeting and pH-responsive charge conversion properties. Our observations indicated that at physiological pH 7.4, P/B + D-MBA@LCNs exhibited a negative charge, while under acidic conditions (pH 5.5, characteristic of the tumor microenvironment), they acquired a positive charge. The particle size of P/B + D-MBA@LCNs was determined to be 168.23 ± 2.66 nm at pH 7.4 and 201.23 ± 1.46 nm at pH 5.5. The crystalline structure of the LCNs was confirmed through small-angle X-ray scattering (SAXS) diffraction patterns. Receptor-mediated endocytosis, facilitated by P/B + D-MBA@LCNs, was confirmed using confocal laser scanning microscopy and flow cytometry. The P/B + D-MBA@LCNs formulation demonstrated a higher rate of G2/M phase arrest (55.20%) compared to free B + D (37.50%) and induced mitochondrial depolarization (59.39%) to a greater extent than P/B + D@LCNs (45.66%). Pharmacokinetic analysis revealed significantly improved area under the curve (AUC) values for both DOX and BPQ when administered as P/B + D-MBA@LCNs, along with enhanced tumor localization. Tumor regression studies exhibited a substantial reduction in tumor size, with P/B + D-MBA@LCNs leading to 3.2- and 1.27-fold reductions compared to B + D and nontargeted P/B + D@LCNs groups, respectively. In summary, this two-tier strategy demonstrates substantial promise for the delivery of a drug combination through the prototype formulation. It offers a potential chemotherapeutic option by minimizing toxic effects on healthy cells while maximizing therapeutic efficacy.
Collapse
Affiliation(s)
- Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | | | - Akhilesh Kumar
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| |
Collapse
|
14
|
Qin M, Xia H, Xu W, Chen B, Wang Y. The spatiotemporal journey of nanomedicines in solid tumors on their therapeutic efficacy. Adv Drug Deliv Rev 2023; 203:115137. [PMID: 37949414 DOI: 10.1016/j.addr.2023.115137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The rapid development of nanomedicines is revolutionizing the landscape of cancer treatment, while effectively delivering them into solid tumors remains a formidable challenge. Currently, there is a huge disconnect on therapeutic response between regulatory approved nanomedicines and laboratory reported nanoparticles. The discrepancy is mainly resulted from the failure of using the classic overall pharmacokinetics behaviors of nanomedicines in tumors to predict the antitumor efficacy. Increasing evidence has revealed that the therapeutic efficacy predominantly relies on the intratumoral spatiotemporal distribution of nanomedicines. This review focuses on the spatiotemporal distribution of systemically administered chemotherapeutic nanomedicines in solid tumor. Firstly, the intratumoral biological barriers that regulate the spatiotemporal distribution of nanomedicines are described in detail. Next, the influences on antitumor efficacy caused by the spatial distribution and temporal drug release of nanomedicines are emphatically analyzed. Then, current methodologies for evaluating the spatiotemporal distribution of nanomedicines are summarized. Finally, the advanced strategies to positively modulate the spatiotemporal distribution of nanomedicines for an optimal tumor therapy are comprehensively reviewed.
Collapse
Affiliation(s)
- Mengmeng Qin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Heming Xia
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Wenhao Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, 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; Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China; Chemical Biology Center, Peking University, Beijing, China.
| |
Collapse
|
15
|
Yadav S. Advanced therapeutics avenues in hepatocellular carcinoma: a novel paradigm. Med Oncol 2023; 40:239. [PMID: 37442842 DOI: 10.1007/s12032-023-02104-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer, and it poses a significant risk to patients health and longevity due to its high morbidity and fatality rates. Surgical ablation, radiotherapy, chemotherapy, and, most recently, immunotherapy have all been investigated for HCC, but none have yielded the desired outcomes. Several unique nanocarrier drug delivery techniques have been studied for their potential therapeutic implications in the treatment of HCC. Nanoparticle-based imaging could be effective for more accurate HCC diagnosis. Since its inception, nanomedicine has significantly transformed the approach to both the treatment and diagnostics of liver cancer. Nanoparticles (NPs) are being studied as a potential treatment for liver cancer because of their ability to carry small substances, such as treatment with chemotherapy, microRNA, and therapeutic genes. The primary focus of this study is on the most current discoveries and practical uses of nanomedicine-based diagnostic and therapeutic techniques for liver cancer. In this section, we had gone over what we know about metabolic dysfunction in HCC and the treatment options that attempt to fix it by targeting metabolic pathways. Furthermore, we propose a multi-target metabolic strategy as a viable HCC treatment option. Based on the findings given here, the scientists believe that smart nanomaterials have great promise for improving cancer theranostics and opening up new avenues for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Plot No.2, Sector 17-A, Yamuna Expressway, Gautam Buddhnagar, Greater Noida, Uttar Pradesh, 201310, India.
| |
Collapse
|
16
|
Kurniawan D, Mathew J, Rahardja MR, Pham HP, Wong PC, Rao NV, Ostrikov KK, Chiang WH. Plasma-Enabled Graphene Quantum Dot Hydrogels as Smart Anticancer Drug Nanocarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206813. [PMID: 36732883 DOI: 10.1002/smll.202206813] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/10/2023] [Indexed: 05/18/2023]
Abstract
One of the major challenges on the way to low-cost, simple, and effective cancer treatments is the lack of smart anticancer drug delivery materials with the requisite of site-specific and microenvironment-responsive properties. This work reports the development of plasma-engineered smart drug nanocarriers (SDNCs) containing chitosan and nitrogen-doped graphene quantum dots (NGQDs) for drug delivery in a pH-responsive manner. Through a customized microplasma processing, a highly cross-linked SDNC with only 4.5% of NGQD ratio can exhibit enhanced toughness up to threefold higher than the control chitosan group, avoiding the commonly used high temperatures and toxic chemical cross-linking agents. The SDNCs demonstrate improved loading capability for doxorubicin (DOX) via π-π interactions and stable solid-state photoluminescence to monitor the DOX loading and release through the Förster resonance energy transfer (FRET) mechanism. Moreover, the DOX loaded SDNC exhibits anticancer effects against cancer cells during cytotoxicity tests at minimum concentration. Cellular uptake studies confirm that the DOX loaded SDNC can be successfully internalized into the nucleus after 12 h incubation period. This work provides new insights into the development of smart, environmental-friendly, and biocompatible nanographene hydrogels for the next-generation biomedical applications.
Collapse
Affiliation(s)
- Darwin Kurniawan
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Jacob Mathew
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Michael Ryan Rahardja
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Hoang-Phuc Pham
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Pei-Chun Wong
- Graduate Institute of Biomedical Optomechatronics, College of Biomedical Engineering, Taipei Medical University, Taipei, 110, Taiwan
| | - Neralla Vijayakameswara Rao
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics and QUT Centre for Materials Science, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia
| | - Wei-Hung Chiang
- Department of Chemical Engineering, National Taiwan University of Science and Technology, Taipei, 10607, Taiwan
| |
Collapse
|
17
|
Kumar Y, Sinha ASK, Nigam KDP, Dwivedi D, Sangwai JS. Functionalized nanoparticles: Tailoring properties through surface energetics and coordination chemistry for advanced biomedical applications. NANOSCALE 2023; 15:6075-6104. [PMID: 36928281 DOI: 10.1039/d2nr07163k] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Significant advances in nanoparticle-related research have been made in the past decade, and amelioration of properties is considered of utmost importance for improving nanoparticle bioavailability, specificity, and catalytic performance. Nanoparticle properties can be tuned through in-synthesis and post-synthesis functionalization operations, with thermodynamic and kinetic parameters playing a crucial role. In spite of robust functionalization techniques based on surface chemistry, scalable technologies have not been explored well. The coordination enhancement via surface functionalization through organic/inorganic/biomolecules material has attracted much attention with morphology modification and shape tuning, which are indispensable aspects in the colloidal phase during biomedical applications. It is envisioned that surface amelioration influences the anchoring properties of nano interfaces for the immobilization of functional groups and biomolecules. In this work, various nanostructure and anchoring methodologies have been discussed, aiming to exploit their full potential in precision engineering applications. Simultaneous discussions on emerging characterization strategies for functionalized assemblies have been made to gain insights into functionalization chemistry. An overview of current advances and prospects of functionalized nanoparticles has been presented, with an emphasis on controllable attributes such as size, shape, morphology, functionality, surface features, Debye and Casimir interactions.
Collapse
Affiliation(s)
- Yogendra Kumar
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai - 600036, India.
| | - A S K Sinha
- Department of Chemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Jais - 229304, India.
| | - K D P Nigam
- Department of Chemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Jais - 229304, India.
- School of Chemical Engineering, University of Adelaide, North Terrace Campus, Adelaide (SA) 5005, Australia
| | - Deepak Dwivedi
- Department of Chemical Engineering, Rajiv Gandhi Institute of Petroleum Technology, Jais - 229304, India.
| | - Jitendra S Sangwai
- Department of Chemical Engineering, Indian Institute of Technology Madras, Chennai - 600036, India.
| |
Collapse
|
18
|
Oladipo AO, Lebelo SL, Msagati TAM. Nanocarrier design–function relationship: The prodigious role of properties in regulating biocompatibility for drug delivery applications. Chem Biol Interact 2023; 377:110466. [PMID: 37004951 DOI: 10.1016/j.cbi.2023.110466] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/14/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
The concept of drug delivery systems as a magic bullet for the delivery of bioactive compounds has emerged as a promising approach in the treatment of different diseases with significant advantages over the limitations of traditional methods. While nanocarrier-based drug delivery systems are the main advocates of drug uptake because they offer several advantages including reduced non-specific biodistribution, improved accumulation, and enhanced therapeutic efficiency; their safety and biocompatibility within cellular/tissue systems are therefore important for achieving the desired effect. The underlying power of "design-interplay chemistry" in modulating the properties and biocompatibility at the nanoscale level will direct the interaction with their immediate surrounding. Apart from improving the existing nanoparticle physicochemical properties, the balancing of the hosts' blood components interaction holds the prospect of conferring newer functions altogether. So far, this concept has been remarkable in achieving many fascinating feats in addressing many challenges in nanomedicine such as immune responses, inflammation, biospecific targeting and treatment, and so on. This review, therefore, provides a diverse account of the recent advances in the fabrication of biocompatible nano-drug delivery platforms for chemotherapeutic applications, as well as combination therapy, theragnostic, and other diseases that are of interest to scientists in the pharmaceutical industries. Thus, careful consideration of the "property of choice" would be an ideal way to realize specific functions from a set of delivery platforms. Looking ahead, there is an enormous prospect for nanoparticle properties in regulating biocompatibility.
Collapse
Affiliation(s)
- Adewale O Oladipo
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida, 1710, South Africa.
| | - Sogolo L Lebelo
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X06, Florida, 1710, South Africa
| | - Titus A M Msagati
- Institute for Nanotechnology and Water Sustainability (iNanoWS), College of Science, Engineering, and Technology, University of South Africa, Private Bag X06, Florida, 1710, South Africa
| |
Collapse
|
19
|
Microcapsule-Based Dose-Dependent Regulation of the Lifespan and Behavior of Adipose-Derived MSCs as a Cell-Mediated Delivery System: In Vitro Study. Int J Mol Sci 2022; 24:ijms24010292. [PMID: 36613737 PMCID: PMC9820487 DOI: 10.3390/ijms24010292] [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: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
The development of “biohybrid” drug delivery systems (DDS) based on mesenchymal stem/stromal cells (MSCs) is an important focus of current biotechnology research, particularly in the areas of oncotheranostics, regenerative medicine, and tissue bioengineering. However, the behavior of MSCs at sites of inflammation and tumor growth is relevant to potential tumor transformation, immunosuppression, the inhibition or stimulation of tumor growth, metastasis, and angiogenesis. Therefore, the concept was formulated to control the lifespan of MSCs for a specific time sufficient for drug delivery to the target tissue by varying the number of internalized microcontainers. The current study addressed the time-dependent in vitro assessment of the viability, migration, and division of human adipose-derived MSCs (hAMSCs) as a function of the dose of internalized polyelectrolyte microcapsules prepared using a layer-by-layer technique. Polystyrene sulfonate (PSS)—poly(allylamine hydrochloride) (PAH)-coated spherical micrometer-sized (diameter ~2−3 µm) vaterite (CaCO3) microcapsules (PAH-PSS)6 with the capping PSS layer were prepared after dissolution of the CaCO3 core template. The Cell-IQ phase contrast imaging results showed that hAMSCs internalized all (PAH-PSS)6 microcapsules saturating the intercellular medium (5−90 particles per cell). A strong (r > 0.7) linear dose-dependent and time-dependent (up to 8 days) regression was observed between the in vitro decrease in cell viability and the number of internalized microvesicles. The approximate time-to-complete-death of hAMSCs at different concentrations of microcapsules in culture was 428 h (1:5 ratio), 339 h (1:10), 252 h (1:20), 247 h (1:45), and 170 h (1:90 ratio). By varying the number of microcontainers loaded into the cells (from 1:10 to 1:90), a dose-dependent exponential decrease in both the movement rate and division rate of hAMSCs was observed. A real-time cell analysis (RTCA) of the effect of (PAH-PSS)6 microcapsules (from 1:5 to 1:20) on hAMSCs also showed a dose- and time-dependent decrease in cell longevity after a 50h study at ratios of 1:10 and 1:20. The incorporation of microcapsules (1:5, 1:20, and 1:45) resulted in a dose-dependent increase in 24−48 h secretion of GRO-α (CXCL1), MIF, and SDF-1α (CXCL12) chemokines in hAMSC culture. In turn, the normalization or inhibition of chemokine secretion occurred after 72 h, except for MIF levels below 5−20 microcapsules, which were internalized by MSCs. Thus, the proposed concept of controlling the lifespan of MSC-based DDS using a dose of internalized PAH-PSS microcapsules could be useful for biomedical applications. (PAH-PSS)6 microcapsule ratios of 1:5 and 1:10 have little effect on the lifespan of hAMSCs for a long time (up to 14−18 days), which can be recommended for regenerative therapy and tissue bioengineering associated with low oncological risk. The microcapsule ratios of 1:20 and 1:45 did not significantly restrict the migratory activity of hAMSCs-based DDS during the time interval required for tissue delivery (up to 4−5 days), followed by cell death after 10 days. Therefore, such doses of microcapsules can be used for hAMSC-based DDS in oncotheranostics.
Collapse
|
20
|
Intelligent nanotherapeutic strategies for the delivery of CRISPR system. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
21
|
Cong VT, Houng JL, Kavallaris M, Chen X, Tilley RD, Gooding JJ. How can we use the endocytosis pathways to design nanoparticle drug-delivery vehicles to target cancer cells over healthy cells? Chem Soc Rev 2022; 51:7531-7559. [PMID: 35938511 DOI: 10.1039/d1cs00707f] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Targeted drug delivery in cancer typically focuses on maximising the endocytosis of drugs into the diseased cells. However, there has been less focus on exploiting the differences in the endocytosis pathways of cancer cells versus non-cancer cells. An understanding of the endocytosis pathways in both cancer and non-cancer cells allows for the design of nanoparticles to deliver drugs to cancer cells whilst restricting healthy cells from taking up anticancer drugs, thus efficiently killing the cancer cells. Herein we compare the differences in the endocytosis pathways of cancer and healthy cells. Second, we highlight the importance of the physicochemical properties of nanoparticles (size, shape, stiffness, and surface chemistry) on cellular uptake and how they can be adjusted to selectively target the dominated endocytosis pathway of cancer cells over healthy cells and to deliver anticancer drug to the target cells. The review generates new thought in the design of cancer-selective nanoparticles based on the endocytosis pathways.
Collapse
Affiliation(s)
- Vu Thanh Cong
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jacinta L Houng
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Maria Kavallaris
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.,Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia.,School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xin Chen
- School of Chemical Engineering and Technology, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, Xi'an Jiao Tong University, Xi'an, China
| | - Richard D Tilley
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia.
| | - J Justin Gooding
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia. .,Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
22
|
Grilli F, Hajimohammadi Gohari P, Zou S. Characteristics of Graphene Oxide for Gene Transfection and Controlled Release in Breast Cancer Cells. Int J Mol Sci 2022; 23:6802. [PMID: 35743245 PMCID: PMC9224565 DOI: 10.3390/ijms23126802] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/16/2022] [Indexed: 12/14/2022] Open
Abstract
Functionalized graphene oxide (GO) nanoparticles are being increasingly employed for designing modern drug delivery systems because of their high degree of functionalization, high surface area with exceptional loading capacity, and tunable dimensions. With intelligent controlled release and gene silencing capability, GO is an effective nanocarrier that permits the targeted delivery of small drug molecules, antibodies, nucleic acids, and peptides to the liquid or solid tumor sites. However, the toxicity and biocompatibility of GO-based formulations should be evaluated, as these nanomaterials may introduce aggregations or may accumulate in normal tissues while targeting tumors or malignant cells. These side effects may potentially be impacted by the dosage, exposure time, flake size, shape, functional groups, and surface charges. In this review, the strategies to deliver the nucleic acid via the functionalization of GO flakes are summarized to describe the specific targeting of liquid and solid breast tumors. In addition, we describe the current approaches aimed at optimizing the controlled release towards a reduction in GO accumulation in non-specific tissues in terms of the cytotoxicity while maximizing the drug efficacy. Finally, the challenges and future research perspectives are briefly discussed.
Collapse
Affiliation(s)
- Francesca Grilli
- Metrology Research Centre, National Research Council of Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6, Canada; (F.G.); (P.H.G.)
- Ottawa-Carleton Institute for Biomedical Engineering, University of Ottawa, 800 King Edward Avenue, Ottawa, ON K1N 6N5, Canada
| | - Parisa Hajimohammadi Gohari
- Metrology Research Centre, National Research Council of Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6, Canada; (F.G.); (P.H.G.)
- Ottawa-Carleton Institute for Biomedical Engineering, University of Ottawa, 800 King Edward Avenue, Ottawa, ON K1N 6N5, Canada
| | - Shan Zou
- Metrology Research Centre, National Research Council of Canada, 100 Sussex Drive, Ottawa, ON K1A 0R6, Canada; (F.G.); (P.H.G.)
- Department of Chemistry, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
23
|
Kumar M, Jha A, Bharti K, Parmar G, Mishra B. Advances in lipid-based pulmonary nanomedicine for the management of inflammatory lung disorders. Nanomedicine (Lond) 2022; 17:913-934. [PMID: 35451334 DOI: 10.2217/nnm-2021-0389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inflammatory lung disorders have become one of the fastest growing global healthcare concerns, with more than 500 million annual cases of disorders such as chronic obstructive pulmonary disease, asthma and pulmonary fibrosis. Owing to environmental changes and socioeconomic disparity, the numbers are expected to grow even more in years to come. The therapeutic strategies and approved drugs currently employed in the management of inflammatory lung disorders show dose-dependent resistance and pharmacokinetic limitations. This review comprehensively discusses lipid-based pulmonary nanomedicine as a potential platform to overcome these barriers while ensuring site-specific drug delivery and minimal side effects in nontargeted tissues for the management of noninfectious inflammatory lung disorders.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| | - Abhishek Jha
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| | - Kanchan Bharti
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| | - Gourav Parmar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India
| |
Collapse
|
24
|
Xuan Y, Gao Y, Guan M, Zhang S. Application of "smart" multifunctional nanoprobes in tumor diagnosis and treatment. J Mater Chem B 2022; 10:3601-3613. [PMID: 35437560 DOI: 10.1039/d2tb00326k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer is one of the major diseases that pose a threat to human health and life, especially because it is difficult to diagnose and cure, and recurs easily. In recent years, the development of nanotechnology has provided researchers with new tools for cancer treatment. In particular, nanoprobes that facilitate integrated diagnosis and treatment, high-resolution imaging, and accurate tumor targeting provide new avenues for the early detection and treatment of cancer. This review focuses on the preparations and applications of two kinds of "smart" multifunctional nanoprobes: "Off-On" nanoprobes and "Charge-Reversal" nanoprobes. This review also briefly discusses their mechanisms of action, as they could provide new ideas for the further development of this field.
Collapse
Affiliation(s)
- Yang Xuan
- Key Laboratory of Biotechnology and Resource Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yating Gao
- Key Laboratory of Biotechnology and Resource Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Meng Guan
- Key Laboratory of Biotechnology and Resource Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Resource Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| |
Collapse
|
25
|
Pang L, Huang X, Zhu L, Xiao H, Li M, Guan H, Gao J, Jin H. [Targeted killing of CD133 + lung cancer stem cells using paclitaxel-loaded PLGA-PEG nanoparticles with CD133 aptamers]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2022; 42:26-35. [PMID: 35249867 DOI: 10.12122/j.issn.1673-4254.2022.01.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To construct a polylactic acid-glycolic acid-polyethylene glycol (PLGA-PEG) nanocarrier (N-Pac-CD133) coupled with a CD133 nucleic acid aptamer carrying paclitaxel for eliminating lung cancer stem cells (CSCs). METHODS Paclitaxel-loaded N-Pac-CD133 was prepared using the emulsion/solvent evaporation method and characterized. CD133+ lung CSCs were separated by magnetic bead separation and identified for their biological behaviors and gene expression profile. The efficiency of paclitaxel-loaded N-Pac-CD133 for targeted killing of lung cancer cells was assessed in vitro. SCID mice were inoculated with A549 cells and received injections of normal saline, empty nanocarrier linked with CD133 aptamer (N-CD133), paclitaxel, paclitaxel-loaded nanocarrier (N-Pac) or paclitaxel-loaded N-Pac-CD133 (n=8, 5 mg/kg paclitaxel) on days 10, 15 and 20, and the tumor weight and body weight of the mice were measured on day 40. RESULTS Paclitaxel-loaded N-Pac-CD133 showed a particle size of about 100 nm with a high encapsulation efficiency (>80%) and drug loading rate (>8%), and was capable of sustained drug release within 48 h. The CD133+ cell population in lung cancer cells showed the characteristic features of lung CSCs, including faster growth rate (30 days, P=0.001) and high expressions of tumor stem cell markers OV6(P < 0.001), CD133 (P=0.001), OCT3/4 (P=0.002), EpCAM (P=0.04), NANOG (P=0.005) and CD44 (P=0.02). Compared with N-Pac and free paclitaxel, paclitaxel-loaded N-Pac-CD133 showed significantly enhanced targeting ability and cytotoxicity against lung CSCs in vitro (P < 0.001) and significantly reduced the formation of tumor spheres (P < 0.001). In the tumor-bearing mice, paclitaxel-loaded N-Pac-CD133 showed the strongest effects in reducing the tumor mass among all the treatments (P < 0.001). CONCLUSION CD133 aptamer can promote targeted delivery of paclitaxel to allow targeted killing of CD133+ lung CSCs. N-Pac-CD133 loaded with paclitaxel may provide an effective treatment for lung cancer by targeting the lung cancer stem cells.
Collapse
Affiliation(s)
- L Pang
- First School of Clinical Medicine, Mudanjiang Medical University, Mudanjiang 157011, China
| | - X Huang
- Department of Respiratory and Critical Care Medicine, Wuhan First Hospital, Wuhan 430022, China
| | - L Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang 157011, China
| | - H Xiao
- Research Department, Mudanjiang Medical University, Mudanjiang 157011, China
| | - M Li
- First School of Clinical Medicine, Mudanjiang Medical University, Mudanjiang 157011, China
| | | | - J Gao
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - H Jin
- Clinical Laboratory, 5Department of Hematology, Affiliated Hongqi Hospital, Mudanjiang Medical University, Mudanjiang 157011, China
| |
Collapse
|
26
|
Dey AK, Nougarède A, Clément F, Fournier C, Jouvin-Marche E, Escudé M, Jary D, Navarro FP, Marche PN. Tuning the Immunostimulation Properties of Cationic Lipid Nanocarriers for Nucleic Acid Delivery. Front Immunol 2021; 12:722411. [PMID: 34497612 PMCID: PMC8419413 DOI: 10.3389/fimmu.2021.722411] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/04/2021] [Indexed: 11/25/2022] Open
Abstract
Nonviral systems, such as lipid nanoparticles, have emerged as reliable methods to enable nucleic acid intracellular delivery. The use of cationic lipids in various formulations of lipid nanoparticles enables the formation of complexes with nucleic acid cargo and facilitates their uptake by target cells. However, due to their small size and highly charged nature, these nanocarrier systems can interact in vivo with antigen-presenting cells (APCs), such as dendritic cells (DCs) and macrophages. As this might prove to be a safety concern for developing therapies based on lipid nanocarriers, we sought to understand how they could affect the physiology of APCs. In the present study, we investigate the cellular and metabolic response of primary macrophages or DCs exposed to the neutral or cationic variant of the same lipid nanoparticle formulation. We demonstrate that macrophages are the cells affected most significantly and that the cationic nanocarrier has a substantial impact on their physiology, depending on the positive surface charge. Our study provides a first model explaining the impact of charged lipid materials on immune cells and demonstrates that the primary adverse effects observed can be prevented by fine-tuning the load of nucleic acid cargo. Finally, we bring rationale to calibrate the nucleic acid load of cationic lipid nanocarriers depending on whether immunostimulation is desirable with the intended therapeutic application, for instance, gene delivery or messenger RNA vaccines.
Collapse
Affiliation(s)
- Arindam K. Dey
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Adrien Nougarède
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Flora Clément
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
- Univ. Grenoble Alpes, CEA, INSERM, IRIG, Biomics, Grenoble, France
| | - Carole Fournier
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Evelyne Jouvin-Marche
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| | - Marie Escudé
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Dorothée Jary
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Fabrice P. Navarro
- Univ. Grenoble Alpes, St Martin d’Hères, France
- CEA, LETI, Division for Biology and Healthcare Technologies, Microfluidic Systems and Bioengineering Lab, Grenoble, France
| | - Patrice N. Marche
- Univ. Grenoble Alpes, St Martin d’Hères, France
- Institute for Advanced Biosciences, Research Center INSERM U1209, CNRS UMR5309, La Tronche, France
| |
Collapse
|
27
|
Nanocarriers as a Tool for the Treatment of Colorectal Cancer. Pharmaceutics 2021; 13:pharmaceutics13081321. [PMID: 34452282 PMCID: PMC8399070 DOI: 10.3390/pharmaceutics13081321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/13/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology is a promising tool for the treatment of cancer. In the past decades, major steps have been made to bring nanotechnology into the clinic in the form of nanoparticle-based drug delivery systems. The great hope of drug delivery systems is to reduce the side effects of chemotherapeutics while simultaneously increasing the efficiency of the therapy. An increased treatment efficiency would greatly benefit the quality of life as well as the life expectancy of cancer patients. However, besides its many advantages, nanomedicines have to face several challenges and hurdles before they can be used for the effective treatment of tumors. Here, we give an overview of the hallmarks of cancer, especially colorectal cancer, and discuss biological barriers as well as how drug delivery systems can be utilized for the effective treatment of tumors and metastases.
Collapse
|
28
|
Zhang X, Zhang M, Wu M, Yang L, Liu R, Zhang R, Zhao T, Song C, Liu G, Zhu Q. Precise Controlled Target Molecule Release through Light-Triggered Charge Reversal Bridged Polysilsesquioxane Nanoparticles. Polymers (Basel) 2021; 13:polym13152392. [PMID: 34371994 PMCID: PMC8346980 DOI: 10.3390/polym13152392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
Precise control of target molecule release time, site, and dosage remains a challenge in controlled release systems. We employed a photoresponsive molecule release system via light-triggered charge reversal nanoparticles to achieve a triggered, stepwise, and precise controlled release platform. This release system was based on photocleavage-bridged polysilsesquioxane nanoparticles which acted as nanocarriers of doxorubicin loaded on the surface via electrostatic interaction. The nanoparticles could reverse into positive charges triggered by 254 nm light irradiation due to the photocleavage of the o-nitrobenzyl bridged segment. The charge reversal property of the nanoparticles could release loaded molecules. Doxorubicin was selected as a positively charged model molecule. The as-prepared nanoparticles with an average size of 124 nm had an acceptable doxorubicin loading content up to 12.8%. The surface charge of the nanoparticles could rapidly reverse from negative (−28.20 mV) to positive (+18.9 mV) upon light irradiation for only 10 min. In vitro release experiments showed a cumulative release up to 96% with continuously enhancing irradiation intensity. By regulating irradiation parameters, precisely controlled drug release was carried out. The typical “stepped” profile could be accurately controlled in an on/off irradiation mode. This approach provides an ideal light-triggered molecule release system for location, timing, and dosage. This updated controlled release system, triggered by near-infrared or infrared light, will have greater potential applications in biomedical technology.
Collapse
|
29
|
Multifunctional polymeric micellar nanomedicine in the diagnosis and treatment of cancer. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 126:112186. [PMID: 34082985 DOI: 10.1016/j.msec.2021.112186] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
Polymeric micelles are a prevalent topic of research for the past decade, especially concerning their fitting ability to deliver drug and diagnostic agents. This delivery system offers outstanding advantages, such as biocompatibility, high loading efficiency, water-solubility, and good stability in biological fluids, to name a few. The multifunctional polymeric micellar architect offers the added capability to adapt its surface to meet the looked-for clinical needs. This review cross-talks the recent reports, proof-of-concept studies, patents, and clinical trials that utilize polymeric micellar family architectures concerning cancer targeted delivery of anticancer drugs, gene therapeutics, and diagnostic agents. The manuscript also expounds on the underlying opportunities, allied challenges, and ways to resolve their bench-to-bedside translation for allied clinical applications.
Collapse
|
30
|
Akkuş-Dağdeviren ZB, Wolf JD, Kurpiers M, Shahzadi I, Steinbring C, Bernkop-Schnürch A. Charge reversal self-emulsifying drug delivery systems: A comparative study among various phosphorylated surfactants. J Colloid Interface Sci 2021; 589:532-544. [PMID: 33493863 DOI: 10.1016/j.jcis.2021.01.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/22/2020] [Accepted: 01/10/2021] [Indexed: 01/04/2023]
Abstract
HYPOTHESIS Phosphorylated surfactants having ethoxylate spacer arms are promising excipients for charge reversal self-emulsifying drug delivery systems (SEDDS). EXPERIMENTS 1,2-Dipalmitoyl-sn-glycero-3-phosphatidic acid disodium salt (PA), 2-((2,3-bis(oleoyloxy)propyl)dimethylammonio)ethyl hydrogen phosphate (DOCP), nonylphenol monophosphate ester (PNPP), C12-15 alcohol 3 ethoxylate phosphate ester (PME) and polyoxyethylene (9) dioctanoyl glycerol pyrophosphate (DGPP) loaded SEDDS were developed and characterized. Zeta potential of SEDDS was measured before and after incubation with intestinal alkaline phosphatase (IAP). Phosphate release was monitored by incubation of SEDDS with isolated as well as cell-associated IAP. Primary amine content on the surface of SEDDS was determined in parallel. Cytotoxicity was evaluated on Caco-2 cells and in vitro hemolysis test was performed. Cellular uptake studies were performed by confocal scanning microscopy. FINDINGS SEDDS formulations exhibited a size in the range of 17 and 193 nm and a polydispersity index (PDI) ≤ 0.5. Charge reversal from negative to positive values could be achieved in case of PNPP and PME loaded SEDDS with a zeta potential changing from -13 mV to +9 mV and from -7 to +2 mV, respectively, within 6 h. Significant amounts of phosphate were released from PNPP and PME loaded SEDDS incubated with isolated IAP and from all formulations incubated with cell-associated IAP in accordance with an increase in primary amines on the surface of oily droplets. SEDDS exhibited a concentration and time-dependent cytotoxicity. PNPP and PME SEDDS displayed an increased cellular uptake.
Collapse
Affiliation(s)
- Zeynep Burcu Akkuş-Dağdeviren
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Julian Dominik Wolf
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; Thiomatrix Forschungs-und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria
| | - Markus Kurpiers
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; Thiomatrix Forschungs-und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria
| | - Iram Shahzadi
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Christian Steinbring
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Andreas Bernkop-Schnürch
- Center for Chemistry and Biomedicine, Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria; Thiomatrix Forschungs-und Beratungs GmbH, Trientlgasse 65, 6020 Innsbruck, Austria.
| |
Collapse
|
31
|
Han QJ, Lan XT, Wen Y, Zhang CZ, Cleary M, Sayyed Y, Huang G, Tuo X, Yi L, Xi Z, Li LY, Zhang QZ. Matrix Metalloproteinase-9-Responsive Surface Charge-Reversible Nanocarrier to Enhance Endocytosis as Efficient Targeted Delivery System for Cancer Diagnosis and Therapy. Adv Healthc Mater 2021; 10:e2002143. [PMID: 33694329 DOI: 10.1002/adhm.202002143] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/16/2021] [Indexed: 12/20/2022]
Abstract
Nanoparticles, that can be enriched in the tumor microenvironment and deliver the payloads into cancer cells, are desirable carriers for theranostic agents in cancer diagnosis and treatment. However, efficient targeted delivery and enhanced endocytosis for probes and drugs in theranostics are still major challenges. Here, a nanoparticle, which is capable of charge reversal from negative to positive in response to matrix metalloproteinase 9 (MMP9) in tumor microenvironment is reported. This nanoparticle is based on a novel charge reversible amphiphilic molecule consisting of hydrophobic oleic acid, MMP9-cleavable peptide, and glutamate-rich segment (named as OMPE). The OMPE-modified cationic liposome forms an intelligent anionic nanohybrid (O-NP) with enhanced endocytosis through surface charge reversal in response to MMP9 in vitro. Successfully, O-NP nanohybrid performs preferential accumulation and enhances the endocytosis in MMP9-expressing xenografted tumors in mouse models, which improve the sensitivity of diagnosis agents and the antitumor effects of drugs in vivo by overcoming their low solubility and/or nonspecific enrichment. These results indicate that O-NP can be a promising delivery platform for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Qiu-Ju Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Xiao-Tong Lan
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Ying Wen
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Chuan-Zeng Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Michael Cleary
- Laboratory Medicine, Yale New Haven Hospital, New Haven, CT, 06510, USA
| | - Yasra Sayyed
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Guangdong Huang
- Sino-science Gene Technology Co., Ltd., Xi'an, Shanxi, 710018, China
| | - Xiaoling Tuo
- Sino-science Gene Technology Co., Ltd., Xi'an, Shanxi, 710018, China
| | - Long Yi
- State Key Laboratory of Organic-Inorganic Composites and Beijing Key Laboratory of Energy Environmental Catalysis, Beijing University of Chemical Technology (BUCT), 15 Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Zhen Xi
- State Key Laboratory of Elemento-Organic Chemistry, National Engineering Research Center of Pesticide (Tianjin), Collaborative Innovation Center of Chemical Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Lu-Yuan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| | - Qiang-Zhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300350, China
| |
Collapse
|
32
|
Rahman M, Almalki WH, Alrobaian M, Iqbal J, Alghamdi S, Alharbi KS, Alruwaili NK, Hafeez A, Shaharyar A, Singh T, Waris M, Kumar V, Beg S. Nanocarriers-loaded with natural actives as newer therapeutic interventions for treatment of hepatocellular carcinoma. Expert Opin Drug Deliv 2021; 18:489-513. [PMID: 33225771 DOI: 10.1080/17425247.2021.1854223] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Cancer has always been a menace for the society. Hepatocellular carcinoma (HCC) is one of the most lethal and 3rdlargest causes of deaths around the world.Area covered: The emergence of natural actives is considered as the greatest boon for fighting cancer. The natural actives take precedence over the traditional chemotherapeutic drugs in terms of their multi-target, multi-level and coordinated effects in the treatment of HCC. Literature reports have indicated the tremendous potential of bioactive natural products in inhibiting the HCC via molecular drug targeting, augmented bioavailability, and the ability for both passive or active targeting and stimulus-responsive drug release characteristics. This review provides a newer treatment approaches involved in the mechanism of action of different natural actives used for the HCC treatment via different molecular pathways. Besides, the promising advantage of natural bioactive-loaded nanocarriers in HCC treatment has also been also presented in this review. Expert opinion: The remarkable outcomes have been observed with therapeutic efficacy of the nanocarriers of natural actives in the treatment of HCC.Furthermore, it requires a thorough assessment of the safety and efficacy evaluation of the nanocarriers for the delivery of targeted natural active ingredients in HCC.].
Collapse
Affiliation(s)
- Mahfoozur Rahman
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-qura University, Saudi Arabia
| | - Majed Alrobaian
- Department of Pharmaceutics & and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Jawed Iqbal
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, Jamia Nagar, New Delhi-110025
| | - Saad Alghamdi
- Laboratory Medicine Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Khalid S Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Nabil K Alruwaili
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Adil Shaharyar
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| | - Tanuja Singh
- Department of Botany, T.P.S College, Patna, Bihar, India
| | - Mohammad Waris
- Department of Botany, T.P.S College, Patna, Bihar, India
| | - Vikas Kumar
- Department of Pharmaceutical Sciences, Shalom Institute of Health & Allied Sciences, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| | - Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Nanomedicine Research Lab, Jamia Hamdard, New Delhi, India
| |
Collapse
|
33
|
Jiang C, Zhao H, Xiao H, Wang Y, Liu L, Chen H, Shen C, Zhu H, Liu Q. Recent advances in graphene-family nanomaterials for effective drug delivery and phototherapy. Expert Opin Drug Deliv 2020; 18:119-138. [PMID: 32729733 DOI: 10.1080/17425247.2020.1798400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Owing to the unique properties of graphene, including large specific surface area, excellent thermal conductivity, and optical absorption, graphene-family nanomaterials (GFNs) have attracted extensive attention in biomedical applications, particularly in drug delivery and phototherapy. AREAS COVERED In this review, we point out several challenges involved in the clinical application of GFNs. Then, we provide an overview of the most recent publications about GFNs in biomedical applications, including diverse strategies for improving the biocompatibility, specific targeting and stimuli-responsiveness of GFNs for drug delivery, codelivery of drug and gene, photothermal therapy, photodynamic therapy, and multimodal combination therapy. EXPERT OPINION Although the application of GFNs is still in the preclinical stage, rational modification of GFNs with functional elements or making full use of GFNs-based multimodal combination therapy might show great potential in biomedicine for clinical application.
Collapse
Affiliation(s)
- Cuiping Jiang
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| | - Haiyue Zhao
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| | - Haiyan Xiao
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| | - Yuan Wang
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| | - Li Liu
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| | - Huoji Chen
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| | - Chunyan Shen
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| | - Hongxia Zhu
- Combining Traditional Chinese and Western Medicine Hospital, Southern Medical University , 510315, Guangzhou, P. R. China
| | - Qiang Liu
- School of Traditional Chinese Medicine, Southern Medical University , Guangzhou, China
| |
Collapse
|
34
|
Zielgerichtete Wirkstoffe für die Krebstherapie: Aktuelle Entwicklungen und Perspektiven. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914511] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
35
|
Chen W, Sun Z, Lu L. Targeted Engineering of Medicinal Chemistry for Cancer Therapy: Recent Advances and Perspectives. Angew Chem Int Ed Engl 2020; 60:5626-5643. [PMID: 32096328 DOI: 10.1002/anie.201914511] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Indexed: 12/13/2022]
Abstract
Severe side effects and poor therapeutic efficacy are the main drawbacks of current anticancer drugs. These problems can be mitigated by targeting, but the targeting efficacy of current drugs is poor and urgently needs improvement. Taking this into consideration, this Review first summarizes the current targeting strategies for cancer therapy in terms of cancer tissue and organelles. Then, we analyse the systematic targeting of anticancer drugs and conclude that a typical journey for a targeted drug administered by intravenous injection is a CTIO cascade of at least four steps. Furthermore, to ensure high overall targeting efficacy, the properties of a targeting drug needed in each step are further analysed, and some guidelines for structure optimization to obtain effective targeting drugs are offered. Finally, some viewpoints highlighting the crucial problems and potential challenges of future research on targeted cancer therapy are presented. This review could actively promote the development of precision medicine against cancer.
Collapse
Affiliation(s)
- Weihua Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, University of Science and Technology of China, Changchun, 130022, China
| | - Zhen Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, University of Science and Technology of China, Changchun, 130022, China
| | - Lehui Lu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, University of Science and Technology of China, Changchun, 130022, China
| |
Collapse
|
36
|
Xu Y, Li X, Gong W, Huang HB, Zhu BW, Hu JN. Construction of Ginsenoside Nanoparticles with pH/Reduction Dual Response for Enhancement of Their Cytotoxicity Toward HepG2 Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:8545-8556. [PMID: 32686932 DOI: 10.1021/acs.jafc.0c03698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The aim of this study is to construct a pH- and reduction-responsive nanodrug delivery system to effectively deliver a ginsenoside (Rh2) and enhance its cytotoxicity against human hepatocarcinoma cells (HepG2). Here, pullulan polysaccharide was grafted by urocanic acid and α-lipoic acid (α-LA) to obtain a copolymer, α-LA-conjugated N-urocanyl pullulan (LA-URPA), which was expected to have pH and redox dual response. Then, the copolymer LA-URPA was used to encapsulate ginsenoside Rh2 to form Rh2 nanoparticles (Rh2 NPs). The results showed that Rh2 NPs exhibited an average size of 119.87 nm with a uniform spherical morphology. Of note, Rh2 NPs showed a high encapsulation efficiency of 86.00%. Moreover, Rh2 NPs possessed excellent pH/reduction dual-responsive drug release under acidic conditions (pH 5.5) and glutathione (GSH) stimulation with a low drug leakage of 14.8% within 96 h. Furthermore, Rh2 NPs with pH/reduction dual response had higher cytotoxicity than Rh2 after incubation with HepG2 cells for 72 h, indicating that Rh2 NPs had a longer circulation time. After the treatment with Rh2 NPs, the excessive increase of reactive oxygen species and the decrease of superoxide dismutase, glutathione (GSH), and mitochondrial membrane potential suggested that the mitochondrial pathway mediated by oxidative stress played a role in this Rh2 NP-induced apoptosis. In conclusion, this study provides a new strategy for improving the application of ginsenoside Rh2 in the food and pharmaceutical fields.
Collapse
Affiliation(s)
- Yu Xu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Xiang Li
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Wei Gong
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Hai-Bo Huang
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Bei-Wei Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, P. R. China
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Jiang-Ning Hu
- National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China
| |
Collapse
|
37
|
Arias-Alpizar G, Kong L, Vlieg RC, Rabe A, Papadopoulou P, Meijer MS, Bonnet S, Vogel S, van Noort J, Kros A, Campbell F. Light-triggered switching of liposome surface charge directs delivery of membrane impermeable payloads in vivo. Nat Commun 2020; 11:3638. [PMID: 32686667 PMCID: PMC7371701 DOI: 10.1038/s41467-020-17360-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/25/2020] [Indexed: 01/14/2023] Open
Abstract
Surface charge plays a fundamental role in determining the fate of a nanoparticle, and any encapsulated contents, in vivo. Herein, we describe, and visualise in real time, light-triggered switching of liposome surface charge, from neutral to cationic, in situ and in vivo (embryonic zebrafish). Prior to light activation, intravenously administered liposomes, composed of just two lipid reagents, freely circulate and successfully evade innate immune cells present in the fish. Upon in situ irradiation and surface charge switching, however, liposomes rapidly adsorb to, and are taken up by, endothelial cells and/or are phagocytosed by blood resident macrophages. Coupling complete external control of nanoparticle targeting together with the intracellular delivery of encapsulated (and membrane impermeable) cargos, these compositionally simple liposomes are proof that advanced nanoparticle function in vivo does not require increased design complexity but rather a thorough understanding of the fundamental nano-bio interactions involved.
Collapse
Affiliation(s)
- Gabriela Arias-Alpizar
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Li Kong
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
- Tongji School of Pharmacy, Huazhong University of Science and Technology, 430030, Wuhan, P.R. China
| | - Redmar C Vlieg
- Leiden Institute of Physics (LION), Leiden University, P.O. Box 9504, 2300, RA, Leiden, The Netherlands
| | - Alexander Rabe
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230, Odense, Denmark
- BioNTech RNA Pharmaceuticals GmbH, An der Goldgrube 12, 55131, Mainz, Germany
| | - Panagiota Papadopoulou
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Michael S Meijer
- Department of Metals in Catalysis, Biomimetics & Inorganic Materials (MCBIM), Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Sylvestre Bonnet
- Department of Metals in Catalysis, Biomimetics & Inorganic Materials (MCBIM), Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands
| | - Stefan Vogel
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5230, Odense, Denmark
| | - John van Noort
- Leiden Institute of Physics (LION), Leiden University, P.O. Box 9504, 2300, RA, Leiden, The Netherlands
| | - Alexander Kros
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands.
| | - Frederick Campbell
- Department o Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, P.O. Box 9502, 2300, RA, Leiden, The Netherlands.
| |
Collapse
|
38
|
Kumar S, Sharma B. Leveraging Electrostatic Interactions for Drug Delivery to the Joint. Bioelectricity 2020; 2:82-100. [PMID: 32856016 DOI: 10.1089/bioe.2020.0014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Arthritis is a debilitating joint disease with a high economic burden and prevalence. There are many challenges delivering therapeutics to the joint, including low bioavailability when administered systemically and low joint retention after intra-articular injection. Therefore, drug delivery systems such as nanoparticles, liposomes, dendrimers, and carrier proteins have been utilized to overcome some of these limitations. To enhance joint tissue localization and retention, there are opportunities to leverage electrostatic interactions between drug carriers and various tissues and cells. These opportunities, as they pertain to specific joint tissues, are explored in this review. Further, the impact that electrostatic interactions has on various drug delivery parameters, such as the formation of a protein corona, the uptake and cytotoxicity, and the biodistribution of the drug delivery systems, is discussed. Lastly, this review summarizes key findings from studies that have investigated the use of electrostatic interactions to increase targeting of specific joint tissues and limitations in preclinical investigations are identified. As more novel targets are discovered in treating arthritis, there will be a continued need to localize therapeutics to specific tissues for greater therapeutic outcomes and hence attention must be paid in designing the drug delivery systems.
Collapse
Affiliation(s)
- Shreedevi Kumar
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Blanka Sharma
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
39
|
Shetab Boushehri MA, Dietrich D, Lamprecht A. Nanotechnology as a Platform for the Development of Injectable Parenteral Formulations: A Comprehensive Review of the Know-Hows and State of the Art. Pharmaceutics 2020; 12:pharmaceutics12060510. [PMID: 32503171 PMCID: PMC7356945 DOI: 10.3390/pharmaceutics12060510] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Within recent decades, the development of nanotechnology has made a significant contribution to the progress of various fields of study, including the domains of medical and pharmaceutical sciences. A substantially transformed arena within the context of the latter is the development and production of various injectable parenteral formulations. Indeed, recent decades have witnessed a rapid growth of the marketed and pipeline nanotechnology-based injectable products, which is a testimony to the remarkability of the aforementioned contribution. Adjunct to the ability of nanomaterials to deliver the incorporated payloads to many different targets of interest, nanotechnology has substantially assisted to the development of many further facets of the art. Such contributions include the enhancement of the drug solubility, development of long-acting locally and systemically injectable formulations, tuning the onset of the drug’s release through the endowment of sensitivity to various internal or external stimuli, as well as adjuvancy and immune activation, which is a desirable component for injectable vaccines and immunotherapeutic formulations. The current work seeks to provide a comprehensive review of all the abovementioned contributions, along with the most recent advances made within each domain. Furthermore, recent developments within the domains of passive and active targeting will be briefly debated.
Collapse
Affiliation(s)
- Maryam A. Shetab Boushehri
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-736428; Fax: +49-228-735268
| | - Dirk Dietrich
- Department of Neurosurgery, University Clinic of Bonn, 53105 Bonn, Germany;
| | - Alf Lamprecht
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- PEPITE EA4267, Institute of Pharmacy, University Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
40
|
|
41
|
Yang F, Xu J, Fu M, Ji J, Chi L, Zhai G. Development of stimuli-responsive intelligent polymer micelles for the delivery of doxorubicin. J Drug Target 2020; 28:993-1011. [PMID: 32378974 DOI: 10.1080/1061186x.2020.1766474] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Doxorubicin is still used as a first-line drug in current therapeutics for numerous types of malignant tumours (including lymphoma, transplantable leukaemia and solid tumour). Nevertheless, to overcome the serious side effects like cardiotoxicity and myelosuppression caused by effective doses of doxorubicin remains as a world-class puzzle. In recent years, the usage of biocompatible polymeric nanomaterials to form an intelligently sensitive carrier for the targeted release in tumour microenvironment has attracted wide attention. These different intelligent polymeric micelles (PMs) could change the pharmacokinetics process of drugs or respond in the special microenvironment of tumour site to maximise the efficacy and reduce the toxicity of doxorubicin in other tissues and organs. Several intelligent PMs have already been in the clinical research stage and planned for market. Therefore, related research remains active, and the latest nanotechnology approaches for doxorubicin delivery are always in the spotlight. Centring on the model drugs doxorubicin, this review summarised the mechanisms of PMs, classified the polymers used in the application of doxorubicin delivery and discussed some interesting and imaginative smart PMs in recent years.
Collapse
Affiliation(s)
- Fan Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jiangkang Xu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Manfei Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Liqun Chi
- Department of Pharmacy, Haidian Maternal and Child Health Hospital of Beijing, Beijing, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
42
|
Zhou Y, Chen X, Cao J, Gao H. Overcoming the biological barriers in the tumor microenvironment for improving drug delivery and efficacy. J Mater Chem B 2020; 8:6765-6781. [PMID: 32315375 DOI: 10.1039/d0tb00649a] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The delivery of drugs to tumors by nanoparticles is a rapidly growing field. However, the complex tumor microenvironment (TME) barriers greatly hinder drug delivery to tumors. In this study, we first summarized the barriers in TME, including anomalous vasculature, rigid extracellular matrix, hypoxia, acidic pH, irregular enzyme level, altered metabolism pathway and immunosuppressive conditions. To overcome these barriers, many strategies have been developed, such as modulating TME, active targeting by ligand modification and biomimetic strategies, and TME-responsive drug delivery strategies to improve nanoparticle penetration, cellular uptake and drug release. Although extensive progress has been achieved, there are still many challenges, which are discussed in the last section. Overall, we carefully discuss the landscape of TME, development for improving drug delivery, and challenges that need to be further addressed.
Collapse
Affiliation(s)
- Yang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610064, China.
| | | | | | | |
Collapse
|
43
|
Fang Z, Pan S, Gao P, Sheng H, Li L, Shi L, Zhang Y, Cai X. Stimuli-responsive charge-reversal nano drug delivery system: The promising targeted carriers for tumor therapy. Int J Pharm 2020; 575:118841. [DOI: 10.1016/j.ijpharm.2019.118841] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 01/04/2023]
|
44
|
Targeted delivery of nanoparticles. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/b978-0-08-102828-5.00010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
45
|
Ma BA, Sun CY. Tumor pH-triggered “charge conversion” nanocarriers with on-demand drug release for precise cancer therapy. J Mater Chem B 2020; 8:9351-9361. [DOI: 10.1039/d0tb01692f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The pHe-triggered “charge conversion” nanocarriers were developed for combined X-ray-induced photodynamic therapy (X-PDT) and hypoxia-activated chemotherapy.
Collapse
Affiliation(s)
- Bo-Ai Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging
- Tianjin Medical University General Hospital
- Tianjin 300052
- P. R. China
- School of Food and Biological Engineering
| | - Chun-Yang Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging
- Tianjin Medical University General Hospital
- Tianjin 300052
- P. R. China
| |
Collapse
|
46
|
Böttger R, Pauli G, Chao PH, AL Fayez N, Hohenwarter L, Li SD. Lipid-based nanoparticle technologies for liver targeting. Adv Drug Deliv Rev 2020; 154-155:79-101. [PMID: 32574575 DOI: 10.1016/j.addr.2020.06.017] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/26/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022]
Abstract
Liver diseases such as hepatitis, cirrhosis, and hepatocellular carcinoma are global health problems accounting for approximately 800 million cases and over 2 million deaths per year worldwide. Major drawbacks of standard pharmacological therapies are the inability to deliver a sufficient concentration of a therapeutic agent to the diseased liver, and nonspecific drug delivery leading to undesirable systemic side effects. Additionally, depending on the specific liver disease, drug delivery to a subset of liver cells is required. In recent years, lipid nanoparticles have been developed to passively and actively target drugs to the liver. The success of this approach has been highlighted by the FDA-approval of the first liver-targeting lipid nanoparticle, ONPATTRO, in 2018 and many other promising candidate technologies are expected to follow. This review summarizes recent developments of various lipid-based liver-targeting technologies, namely solid-lipid nanoparticles, liposomes, niosomes and micelles, and discusses the challenges and future perspectives in this field.
Collapse
|
47
|
Li D, Qin J, Sun M, Yan G, Tang R. pH-sensitive, dynamic graft polymer micelles via simple synthesis for enhanced chemotherapeutic efficacy. J Biomater Appl 2019; 34:1059-1070. [DOI: 10.1177/0885328219894695] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To promote chemotherapeutic efficacy and easier clinical transformation, a series of pH-sensitive and dynamic drug delivery systems with facile two-step synthesis and simple structure have been successfully constructed by the tunable grafting reaction between pH-sensitive ortho ester and poly(vinyl alcohol). The amphipathic graft macromolecules (PVA- g-OE x, x represents the percentage of feed between ortho esters and hydroxyl groups of polyvinyl alcohol) could self-assemble into micelles and doxorubicin was embedded. These micelles exhibited pH-sensitivity to both extracellular and intracellular pH and demonstrated the following characteristics: (i) maintaining long-term storage and blood circulation stability at pH 7.4; (ii) responding to tumoral extracellular pH value following gradually larger nanoparticles for improved drug accumulation and retention; (iii) being sensitive to tumoral intracellular pH value following disintegration for rapid drug release to improve toxicity to tumor cells. Moreover, the doxorubicin-loaded micelle (PVA- g-OE30-DOX) showed similar cytotoxicity to free doxorubicin in vitro, but stronger tumor penetration and inhibition ability in vitro human liver carcinoma cell line multicellular tumor spheroids. In vivo biodistribution and tumor inhibition examinations demonstrated that PVA- g-OE30-DOX had more superior efficacy in significantly enhancing drug accumulation in tumor, restraining tumor growth while decreasing drug concentration in normal tissues. The pH-sensitive, dynamic graft polymer micelles via simple synthesis could be considered as a promising and effective drug carrier in tumor therapy.
Collapse
Affiliation(s)
- Dapeng Li
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Jiejie Qin
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Min Sun
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Guoqing Yan
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Rupei Tang
- Engineering Research Center for Biomedical Materials, School of Life Science, Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| |
Collapse
|
48
|
Li C, Wang J, Wang Y, Gao H, Wei G, Huang Y, Yu H, Gan Y, Wang Y, Mei L, Chen H, Hu H, Zhang Z, Jin Y. Recent progress in drug delivery. Acta Pharm Sin B 2019; 9:1145-1162. [PMID: 31867161 PMCID: PMC6900554 DOI: 10.1016/j.apsb.2019.08.003] [Citation(s) in RCA: 455] [Impact Index Per Article: 75.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 01/05/2023] Open
Abstract
Drug delivery systems (DDS) are defined as methods by which drugs are delivered to desired tissues, organs, cells and subcellular organs for drug release and absorption through a variety of drug carriers. Its usual purpose to improve the pharmacological activities of therapeutic drugs and to overcome problems such as limited solubility, drug aggregation, low bioavailability, poor biodistribution, lack of selectivity, or to reduce the side effects of therapeutic drugs. During 2015-2018, significant progress in the research on drug delivery systems has been achieved along with advances in related fields, such as pharmaceutical sciences, material sciences and biomedical sciences. This review provides a concise overview of current progress in this research area through its focus on the delivery strategies, construction techniques and specific examples. It is a valuable reference for pharmaceutical scientists who want to learn more about the design of drug delivery systems.
Collapse
Affiliation(s)
- Chong Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jiancheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiguang Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Huile Gao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai 201203, China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haijun Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yong Gan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yongjun Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lin Mei
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou 510275, China
| | - Huabing Chen
- School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhiping Zhang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| |
Collapse
|
49
|
Ferroni C, Del Rio A, Martini C, Manoni E, Varchi G. Light-Induced Therapies for Prostate Cancer Treatment. Front Chem 2019; 7:719. [PMID: 31737599 PMCID: PMC6828976 DOI: 10.3389/fchem.2019.00719] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PC) is one of the most widespread tumors affecting the urinary system and the fifth-leading cause from cancer death in men worldwide. Despite PC mortality rates have been decreasing during the last years, most likely due to an intensification of early diagnosis, still more than 300,000 men die each year because of this disease. In this view, researchers in all countries are engaged in finding new ways to tackle PC, including the design and synthesis of novel molecular and macromolecular entities able to challenge different PC biological targets, while limiting the extent of unwanted side effects that significantly limit men's life quality. Among this field of research, photo-induced therapies, such as photodynamic and photothermal therapies (PDT and PTT), might represent an important advancement in PC treatment due to their extremely localized and controlled cytotoxic effect, as well as their low incidence of side effects and tumor resistance occurrence. Based on these considerations, this review aims to gather and discuss the last 5-years literature reports dealing with the synthesis and biological activity of molecular conjugates and nano-platforms for photo-induced therapies as co-adjuvant or combined therapeutic modalities for the treatment of localized PC.
Collapse
Affiliation(s)
- Claudia Ferroni
- Institute of Organic Synthesis and Photoreactivity – ISOF, Italian National Research Council, Bologna, Italy
| | - Alberto Del Rio
- Institute of Organic Synthesis and Photoreactivity – ISOF, Italian National Research Council, Bologna, Italy
- Innovamol Consulting Srl, Modena, Italy
| | - Cecilia Martini
- Institute of Organic Synthesis and Photoreactivity – ISOF, Italian National Research Council, Bologna, Italy
| | - Elisabetta Manoni
- Institute of Organic Synthesis and Photoreactivity – ISOF, Italian National Research Council, Bologna, Italy
| | - Greta Varchi
- Institute of Organic Synthesis and Photoreactivity – ISOF, Italian National Research Council, Bologna, Italy
| |
Collapse
|
50
|
Mohammed F, Ke W, Mukerabigwi JF, M Japir AAWM, Ibrahim A, Wang Y, Zha Z, Lu N, Zhou M, Ge Z. ROS-Responsive Polymeric Nanocarriers with Photoinduced Exposure of Cell-Penetrating Moieties for Specific Intracellular Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2019; 11:31681-31692. [PMID: 31397163 DOI: 10.1021/acsami.9b10950] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In situ modulation of the surface properties on the micellar drug delivery nanocarriers offers an efficient method to improve the drug delivery efficiency into cells while maintaining stealth and stability during blood circulation. Light has been demonstrated to be a temporally and spatially controllable tool to improve cellular internalization of nanoparticles. Herein, we develop reactive oxygen species (ROS)-responsive mixed polymeric micelles with photoinduced exposure of cell-penetrating moieties via photodynamic ROS production, which can facilitate cellular internalization of paclitaxel (PTX) and chlorin e6 (Ce6)-coloaded micelles for the synergistic effect of photodynamic and chemotherapy. The thioketal-bond-linked block polymers poly(ε-caprolactone)-TL-poly(N,N-dimethylacrylamide) (PCL-TL-PDMA) with a long PDMA block are used to self-assemble into mixed micelles with PCL-b-poly(2-guanidinoethyl methacrylate) (PCL-PGEMA) consisting of a short PGEMA block, which are further used to coencapsulate PTX and Ce6. After intravenous injection, prolonged blood circulation of the micelles guarantees high tumor accumulation. Upon irradiation by 660 nm light, ROS production of the micelles by Ce6 induces cleavage of PDMA to expose PGEMA shells for significantly improved cellular internalization. The combination of photodynamic therapy and chemotherapy inside the tumor cells achieves improved antitumor efficacy. The design of ROS-responsive mixed polymeric nanocarriers represents a novel and efficient approach to realize both long blood circulation and high-efficiency cellular internalization for combined photodynamic and chemotherapy under light irradiation.
Collapse
Affiliation(s)
- Fathelrahman Mohammed
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| | - Wendong Ke
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| | - Jean Felix Mukerabigwi
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| | - Abd Al-Wali Mohammed M Japir
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| | - Alhadi Ibrahim
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| | - Yuheng Wang
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| | - Zengshi Zha
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| | - Nannan Lu
- Department of Oncology , The First Affiliated Hospital of University of Science and Technology of China , Hefei 230001 , Anhui , China
| | - Min Zhou
- Neurocritical Care Unit, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine , University of Science and Technology of China , Hefei 230001 , Anhui , China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering , University of Science and Technology of China , Hefei 230026 , Anhui , China
| |
Collapse
|