1
|
Lin M, Liu D, Gong Y, Shu L, Wang H, Zhang G, Li J, Gao Z, Sun J, Chen X. Bioactive Assembly Cofactor-Assisted Ursolic Acid Helix for Enhanced Anticancer Efficacy via In Situ Virus-like Transition. J Am Chem Soc 2025; 147:17010-17021. [PMID: 40354555 DOI: 10.1021/jacs.5c01214] [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: 05/14/2025]
Abstract
Natural bioactive pentacyclic triterpenoids, such as ursolic acid (UA), hold significant potential as anticancer agents. However, their clinical application is limited by their poor solubility and bioavailability. Herein, we developed a novel polypeptoid assembly cofactor-assisted nanoplatform designed to enhance UA's therapeutic efficacy through in situ self-assembly within the tumor microenvironment (TME). Bioactive polypeptoid polyelectrolytes, inspired by natural molecular chaperones, were utilized as assembly cofactors to guide UA's co-assembly into stimuli-responsive nanostructures. These polypeptoids provide precise control over the assembly process, improving stability and enabling reversible, pH-responsive transformations. Acid-responsive groups and the target molecule lactobionic acid further promote the specificity and efficacy of UA delivery. Under neutral conditions, the assemblies retain a helical fibrous structure, while in the acidic TME, they transform into virus-like clusters composed of assembly subunits, facilitating deeper tumor penetration. Once internalized, these nanoparticles escape into the cytoplasm and accumulate around the mitochondria, where the oxidation of thioether bonds triggers the release of UA and polypeptoids, causing mitochondrial damage and apoptosis. Some nanoparticles reassemble into fibrous structures intracellularly, extending their retention in tumor cells and potentially leading to mitochondria damage. Notably, the nanoplatform demonstrates excellent synergistic effects, achieving significantly higher therapeutic efficiency compared with individual components, including UA and polypeptoids. In vivo studies further confirmed the effectiveness, demonstrating significant tumor growth suppression and reduced metastasis. By integrating the therapeutic UA with bioactive polypeptoids under precise control, this synergistic platform represents a highly efficient and targeted approach to cancer therapy, offering a promising new opportunity for natural compounds for advanced nanomedicine.
Collapse
Affiliation(s)
- Min Lin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Dandan Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Yiyu Gong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Lilei Shu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Helin Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Guojing Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Jiayi Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Zixin Gao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Jing Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
| | - Xuesi Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 130012 Changchun, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022 Changchun, China
| |
Collapse
|
2
|
Li Y, Sun H, Cao D, Guo Y, Wu D, Yang M, Wang H, Shao X, Li Y, Liang Y. Overcoming Biological Barriers in Cancer Therapy: Cell Membrane-Based Nanocarrier Strategies for Precision Delivery. Int J Nanomedicine 2025; 20:3113-3145. [PMID: 40098719 PMCID: PMC11913051 DOI: 10.2147/ijn.s497510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Given the unique capabilities of natural cell membranes, such as prolonged blood circulation and homotypic targeting, extensive research has been devoted to developing cell membrane-inspired nanocarriers for cancer therapy, while most focused on overcoming one or a few biological barriers. In fact, the journey of nanosystems from systemic circulation to tumor cells involves intricate processes, encompassing blood circulation, tissue accumulation, cancer cell targeting, endocytosis, endosomal escape, intracellular trafficking to target sites, and therapeutic action, all of which pose limitations to their clinical translation. This underscores the necessity of meticulously considering these biological barriers in the design of cell membrane-mimetic nanocarriers. In this review, we delineate the functions and applications of diverse types of cell membranes in nanocarrier systems. We elaborate on the biological hurdles encountered at each stage of the biomimetic nanoparticle's odyssey to the target, and comprehensively discuss the obstacles imposed by the tumor microenvironment for precise delivery. Subsequently, we systematically review contemporary cell membrane-based strategies aimed at overcoming these multi-level biological barriers, encompassing hybrid cell membrane (HCM) camouflage, tumor microenvironment remodeling, endosomal/lysosomal escape, multidrug resistance (MDR) reversal, optimization of nanoparticle physicochemical properties, and so on. Finally, we outline potential strategies to accelerate the development of cell membrane-inspired precision nanocarriers and discuss the challenges that must be addressed to enhance their clinical applicability. This review serves as a guide for refining the study of cell membrane-mimetic nanosystems in surmounting in vivo delivery barriers, thereby significantly contributing to advancing the development and application of cell membrane-based nanoparticles in cancer delivery.
Collapse
Affiliation(s)
- Yuping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dianchao Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dongyang Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Menghao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hongming Wang
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Xiaowei Shao
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| |
Collapse
|
3
|
Chen XQ, Cui SS, Chen YZ, Wang CY, Liu Q, Qi YK, Du SS. Efficient Delivery of Oncolytic Peptide LTX-315 by ZIF-8: pH-Responsive Release, Improved Stability, and Reduced Hemolysis. Mol Pharm 2025; 22:1449-1461. [PMID: 39913295 DOI: 10.1021/acs.molpharmaceut.4c01144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
The first-in-class oncolytic peptide LTX-315 has exhibited positive anticancer responses in multiple phase I/II clinical trials. Nevertheless, the linear peptide LTX-315 suffers from poor proteolytic stability and undesired toxicity, especially hemolysis, which may limit its widespread applications. Except for the direct structural modifications, drug delivery systems (DDSs) are expected to protect LTX-315 from degradation and shield its hemolytic properties. Therefore, the LTX-315 and zeolitic imidazolate framework (ZIF-8)-based nanoparticles (NPs) were constructed with a high LTX-315 encapsulation rate of 59.9%, utilizing the biomineralized "one-pot method" in an aqueous system. The release of LTX-315, in vitro anticancer potency, serum stability, anticancer durability, antimigration activity, hemolysis effect, subcellular localization, and the membrane disruption/permeation effects of LTX-315@ZIF-8 NPs were investigated. LTX-315@ZIF-8 NPs exhibited potent cytotoxicity against cancer cells. The serum stability experiment and time-inhibition curve assay indicated that ZIF-8 NPs could effectively improve the stability of LTX-315, prolong the duration of anticancer action, and enhance the cytostatic potency. Especially, the LTX-315@ZIF-8 NPs not only effectively attenuated the hemolytic toxicity of LTX-315 but also achieved the pH-responsive release of LTX-315. The mechanism investigation indicated that LTX-315@ZIF-8 NPs possessed potent membranolytic activity and reduced the mitochondrial membrane potential to trigger cell death. Collectively, this paper not only established a robust strategy to improve the stability and reduce the hemolytic properties of LTX-315 but also provided a reliable reference for the future delivery of oncolytic peptides.
Collapse
Affiliation(s)
- Xin-Qi Chen
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Su-Su Cui
- Suzhou Jinchang Street Bailian Community Health Service Center, Suzhou 215000, China
| | - Yu-Zhen Chen
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Cai-Yun Wang
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Qing Liu
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yun-Kun Qi
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Shan-Shan Du
- State Key Laboratory Base for Eco-Chemical Engineering in College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
- Department of Natural Medicinal Chemistry and Pharmacognosy, School of Pharmacy, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| |
Collapse
|
4
|
Guo Z, Huang T, Lv X, Yin R, Wan P, Li G, Zhang P, Xiao C, Chen X. Tumor microenvironment-activated polypeptide nanoparticles for oncolytic immunotherapy. Biomaterials 2025; 314:122870. [PMID: 39369669 DOI: 10.1016/j.biomaterials.2024.122870] [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: 07/31/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Cationic oncolytic polypeptides have gained increasing attention owing to their ability to directly lyse cancer cells and activate potent antitumor immunity. However, the low tumor cell selectivity and inherent toxicity induced by positive charges of oncolytic polypeptides hinder their systemic application. Herein, a tumor microenvironment-responsive nanoparticle (DNP) is developed by the self-assembly of a cationic oncolytic polypeptide (PLP) with a pH-sensitive anionic polypeptide via electrostatic interactions. After the formation of DNP, the positive charges of PLP are shielded. DNPs can keep stable in physiological conditions (pH 7.4) but respond to acidic tumor microenvironment (pH 6.8) to release oncolytic PLP. As a result, DNPs evoke potent immunogenic cell death by disrupting cell membranes, damaging mitochondria and increasing intracellular levels of reactive oxygen species. In vivo results indicate that DNPs significantly improve the biocompatibility of PLP, and inhibit tumor growth, recurrence and metastasis by direct oncolysis and activation of antitumor immune responses. In summary, these results indicate that pH-sensitive DNPs represent a prospective strategy to improve the tumor selectivity and biosafety of cationic polymers for oncolytic immunotherapy.
Collapse
Affiliation(s)
- Zhihui Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Tianze Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Xueli Lv
- College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Renyong Yin
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Pengqi Wan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Gao Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, 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; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| |
Collapse
|
5
|
Delgado-Pujol EJ, Martínez G, Casado-Jurado D, Vázquez J, León-Barberena J, Rodríguez-Lucena D, Torres Y, Alcudia A, Begines B. Hydrogels and Nanogels: Pioneering the Future of Advanced Drug Delivery Systems. Pharmaceutics 2025; 17:215. [PMID: 40006582 PMCID: PMC11859140 DOI: 10.3390/pharmaceutics17020215] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Conventional drug delivery approaches, including tablets and capsules, often suffer from reduced therapeutic effectiveness, largely attributed to inadequate bioavailability and difficulties in ensuring patient adherence. These challenges have driven the development of advanced drug delivery systems (DDS), with hydrogels and especially nanogels emerging as promising materials to overcome these limitations. Hydrogels, with their biocompatibility, high water content, and stimuli-responsive properties, provide controlled and targeted drug release. This review explores the evolution, properties, and classifications of hydrogels versus nanogels and their applications in drug delivery, detailing synthesis methods, including chemical crosslinking, physical self-assembly, and advanced techniques such as microfluidics and 3D printing. It also examines drug-loading mechanisms (e.g., physical encapsulation and electrostatic interactions) and release strategies (e.g., diffusion, stimuli-responsive, and enzyme-triggered). These gels demonstrate significant advantages in addressing the limitations of traditional DDS, offering improved drug stability, sustained release, and high specificity. Their adaptability extends to various routes of administration, including topical, oral, and injectable forms, while emerging nanogels further enhance therapeutic targeting through nanoscale precision and stimuli responsiveness. Although hydrogels and nanogels have transformative potential in personalized medicine, challenges remain in scalable manufacturing, regulatory approval, and targeted delivery. Future strategies include integrating biosensors for real-time monitoring, developing dual-stimuli-responsive systems, and optimizing surface functionalization for specificity. These advancements aim to establish hydrogels and nanogels as cornerstones of next-generation therapeutic solutions, revolutionizing drug delivery, and paving the way for innovative, patient-centered treatments.
Collapse
Affiliation(s)
- Ernesto J. Delgado-Pujol
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
- Departamento de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, Universidad de Sevilla, 41011 Sevilla, Spain;
| | - Guillermo Martínez
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| | - David Casado-Jurado
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Juan Vázquez
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - Jesús León-Barberena
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, 41012 Sevilla, Spain;
| | - David Rodríguez-Lucena
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| | - Yadir Torres
- Departamento de Ingeniería y Ciencia de los Materiales y del Transporte, Escuela Politécnica Superior, Universidad de Sevilla, 41011 Sevilla, Spain;
| | - Ana Alcudia
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| | - Belén Begines
- Departamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (E.J.D.-P.); (G.M.); (D.C.-J.); (D.R.-L.); (A.A.)
| |
Collapse
|
6
|
Zhu H, Xu C, Geng Y, Shen Y, Qiu N. Endoplasmic Reticulum-Targeted Polymer-Manganese Nanocomplexes for Tumor Immunotherapy. ACS NANO 2025; 19:4959-4972. [PMID: 39854168 DOI: 10.1021/acsnano.4c17279] [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: 01/26/2025]
Abstract
Manganese ions (Mn2+) are an immune activator that enhances the activation of both cGAS and STING proteins. The STING signaling activation and subsequential immune responses are predominantly associated with endoplasmic reticulum (ER). Therefore, ER targeting of Mn2+ in the subcellular compartments would promote the activation of STING signaling pathways. Herein, we report the design of ER-targeted manganese-based nanocomplexes (NCs) by complexation of Mn2+ with a zwitterionic polymer, poly[2-(N-oxide-N,N-dimethylamino) ethyl methacrylate] (OPDMA). The Mn/OPDMA nanocomplexes (Mn/OPDMA NCs) keep a long blood circulation for tumor accumulation and trigger adsorption-mediated transcytosis for extravasation and deep tumor penetration. Notably, in the tumor-associated macrophages, the Mn/OPDMA NCs can preferentially translocate to their ERs, significantly enhancing cGAS-STING pathway activation for tumor-associated macrophage polarization and IFN-β secretion. In mouse colon and hepatocellular cancer models, the intravenously administrated Mn/OPDMA NCs efficiently remodel tumor immune microenvironment, greatly retard tumor growths by 2.4- to 5-fold, and prolong the mouse survivals compared to free Mn2+-treated mice. This study provides the ER-targeted delivery of Mn2+ that achieves robust STING activation and, thus, potent systemic tumor inhibition without the toxicity of free Mn2+.
Collapse
Affiliation(s)
- Haoru Zhu
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education of China, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Chang Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yu Geng
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education of China, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education of China, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| |
Collapse
|
7
|
Khan AH, Basak A, Zaman A, Das PK. Inherently targeted estradiol-derived carbon dots for selective killing of ER (+) breast cancer cells via oridonin-triggered p53 pathway activation. J Mater Chem B 2024; 12:11708-11720. [PMID: 39435655 DOI: 10.1039/d4tb01415d] [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: 10/23/2024]
Abstract
One of the most prevalent cancers globally is breast cancer and approximately two thirds of the breast cancers are hormone receptor positive with estrogen receptors (ER) being a prominent target. Notably, p53 that controls several cellular functions and prevents tumor formation, gets suppressed in breast cancers. Reactivation of p53 can lead to cell cycle arrest as well as apoptosis. Therefore, targeting the estrogen receptor for selective delivery of anticancer drugs that can reactivate p53 in ER (+) breast cancers can be a crucial method in breast cancer therapy. Herein, we have designed and developed estradiol-derived inherently targeted specific carbon dots (E2-CA-CD) from 17β-estradiol and citric acid following a solvothermal method. The synthesized carbon dots were characterized using spectroscopic and microscopic techniques. The water soluble, intrinsically fluorescent E2-CA-CD showed excellent biocompatibility in MCF-7, MDA-MB-231 as well as NIH3T3 cells and demonstrated target specific bioimaging in ER (+) MCF-7 cells due to the overexpressed ER receptors. Furthermore, oridonin, a well-known hydrophobic anticancer drug capable of upregulating the p53 pathway, was loaded on the carbon dots to increase its bioavailability. E2-CA-CD-Ori caused ∼2.2 times higher killing in ER (+) MCF-7 cells compared to ER (-) MDA-MB-231 cells and normal cells NIH3T3. Also, E2-CA-CD-Ori showed ∼3 fold better killing in MCF-7 cells compared to native oridonin. E2-CA-CD-Ori-induced killing of MCF-7 cells took place through the early to late apoptotic pathway along with the elevation of the intracellular ROS level. Importantly, E2-CA-CD-Ori triggered the activation of the p53 pathway in MCF-7 cells, which in turn induced apoptosis involving the upregulation of Bax and downregulation of Bcl-2 leading to the selective and efficient killing of ER (+) MCF-7 cells.
Collapse
Affiliation(s)
- Aftab Hossain Khan
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Ambalika Basak
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Afreen Zaman
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| | - Prasanta Kumar Das
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata - 700 032, India.
| |
Collapse
|
8
|
Ma X, Lin N, Hu K, Xu C, Yang Q, Feng Y, Liu P, Ding H, Xu M, Shi Q, Chen H, Xue F. An acid-activatable fluorouracil prodrug for colorectal cancer synergistic therapy. Acta Biomater 2024; 185:350-360. [PMID: 39013485 DOI: 10.1016/j.actbio.2024.07.012] [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: 04/11/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024]
Abstract
5-Fluorouracil has demonstrated certain efficiency in patients with colorectal cancer. However, significant side effects of use by injection are common. To address this issue defects, a reengineered 5'-deoxy-5-fluorocytidine (DFCR) based drug delivery system (POACa) is developed as a prominent tumor-selective nano-activator. Investigations demonstrate that the constructed nano-activator exhibits good biocompatibility and high therapeutic efficiency in mice with subcutaneous and orthotopic SW-480 colorectal tumors, as its activity is strictly dependent on the tumor-associated acid environment and thymidine phosphorylase. These strategies diminish the off-target toxicity and improve the specificity and sensitivity of human colorectal cancer cells to 5-Fu, obtaining potent efficiency by the combination of H2O2 mediated oxidative stress, calcium overload and 5-Fu-induced chemotherapy (the combination index is 0.11). Overall, the engineered nano-activator exhibits a high therapeutic index in vitro and in vivo. STATEMENT OF SIGNIFICANCE: In this study, we designed and prepared a pH-responsive polymer to synchronously deliver DFCR (5'-deoxy-5-fluorocytidine, a prodrug of 5-Fu), Ca2+ and H2O2. The constructed nano-activator was denoted as POACa. (1) To address the problem of premature leakage of cargo by physical embedding, our research modified the inactive prodrug DFCR through chemical bonding. (2) The activation of the prepared nano-activator was strictly dependent on the tumor-associated acid environment and thymidine phosphorylase, providing the drug delivery system with inherent safety. (3) A distinctly low combination index value (0.11) of CaO2 and DFCR indicated that POACa has a prominent tumor suppression effect by tumor calcium overload sensitized chemotherapy and H2O2 mediated cytotoxicity.
Collapse
Affiliation(s)
- Xiaoqian Ma
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Nuo Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Ke Hu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Chao Xu
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian Province, China. Clinical Medical Center for Digestive Diseases, Fujian Provincial Hospital, China
| | - Qing Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Yushuo Feng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Peifei Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Haizhen Ding
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Mengjiao Xu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Qianqian Shi
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China
| | - Hongmin Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, China. State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Intergration in Vaccine Research, Xiamen University, China.
| | - Fangqin Xue
- Department of Gastrointestinal Surgery, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, Fujian Province, China. Clinical Medical Center for Digestive Diseases, Fujian Provincial Hospital, China.
| |
Collapse
|
9
|
Kolay S, Das M, Mondal A, Sengupta A, Bag S, De P, Molla MR. Enzyme-Triggered Degradation of Supramolecularly Cross-Linked Polymersomes of Azobenzene-Based Polyurethane: Cell-Selective Anticancer Drug Release. Biomacromolecules 2024; 25:5068-5080. [PMID: 39041235 DOI: 10.1021/acs.biomac.4c00485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Enzyme-responsive self-assembled nanostructures for drug delivery applications have gained a lot of attention, as enzymes exhibit dysregulation in many disease-associated microenvironments. Azoreductase enzyme levels are strongly elevated in many tumor tissues; hence, here, we exploited the altered enzyme activity of the azoreductase enzyme and designed a main-chain azobenzene-based amphiphilic polyurethane, which self-assembles into a vesicular nanostructure and is programmed to disassemble in response to a specific enzyme, azoreductase, with the help of the nicotinamide adenine dinucleotide phosphate (NADPH) coenzyme in the hypoxic environment of solid tumors. The vesicular nanostructure sequesters, stabilizes the hydrophobic anticancer drug, and releases the drug in a controlled fashion in response to enzyme-triggered degradation of azo-bonds and disruption of vesicular assembly. The biological evaluation revealed tumor extracellular matrix pH-induced surface charge modulation, selective activated cellular uptake to azoreductase overexpressed lung cancer cells (A549), and the release of the anticancer drug followed by cell death. In contrast, the benign nature of the drug-loaded vesicular nanostructure toward normal cells (H9c2) suggested excellent cell specificity. We envision that the main-chain azobenzene-based polyurethane discussed in this manuscript could be considered as a possible selective chemotherapeutic cargo against the azoreductase overexpressed cancer cells while shielding the normal cells from off-target toxicity.
Collapse
Affiliation(s)
- Soumya Kolay
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Madhuchhanda Das
- Department of Life Science & Biotechnology, Jadavpur University, 188, Raja S. C. M Road, Kolkata 700032, India
| | - Arun Mondal
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| | - Arunima Sengupta
- Department of Life Science & Biotechnology, Jadavpur University, 188, Raja S. C. M Road, Kolkata 700032, India
| | - Sagar Bag
- Department of Chemical Science, Indian Institute of Science Education and Research, Mohanpur, Nadia, Kolkata, West Bengal 741246, India
| | - Priyadarsi De
- Department of Chemical Science, Indian Institute of Science Education and Research, Mohanpur, Nadia, Kolkata, West Bengal 741246, India
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata 700009, India
| |
Collapse
|
10
|
Yang Y, Wang S, Liu L, Yue B, Qi P, Zhang M, Song S. A Triterpene-Based bioactive drug delivery system for combined chemotherapy of liver cancer. Eur J Pharm Biopharm 2024; 201:114378. [PMID: 38917949 DOI: 10.1016/j.ejpb.2024.114378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/05/2024] [Accepted: 06/20/2024] [Indexed: 06/27/2024]
Abstract
Carrier materials always account for the majority particularly in nanosized formulations, which are administrated along with the active ingredient part might result in metabolism related toxicity. The usage of bioactive excipients could not only reduce the sided effect but also provide additional therapeutic effects. In the present study, a triterpene based micellar drug delivery system was developed using a bioactive solanesol derivative. Solanesylamine was prepared firstly followed by conjugating with poly (ethylene glycol) using maleic acid amide linkage. The amphiphilic drug carrier PEGylated (2-propyl-3-methylmaleic acid)-block-solanesol amine (mPEG-CDM-NH-SOL) could be formed into micelles and loaded with doxorubicin (DOX) inside. The micelles were about 112 nm in size and the drug loading content was about 5.97 wt%. An acid triggered drug release behavior was obviously observed for the DOX loaded pH-sensitive micelle mPEG-CDM-NH-SOL-DOX. While not for DOX-loaded micelles without pH-sensitivity (mPEG-NHS-NH-SOL). CCK8 assay showed that the micelles of PEGylated solanesylamines exhibited certain inhibitory effect on tumor cells at high concentration and the pH sensitive ones seemed more toxic. In vivo studies showed that the pH sensitive mPEG-CDM-NH-SOL-DOX had a superior anti-tumor effect, indicating its great potential in cancer treatment.
Collapse
Affiliation(s)
- Yanwei Yang
- Department of Pharmacy, the First Affiliated Hospital of Henan University, Kaifeng 475004, China
| | - Shuaichao Wang
- School of Pharmacy, Henan University, Kaifeng, China 475004
| | - Lei Liu
- School of Pharmacy, Henan University, Kaifeng, China 475004.
| | - Bolin Yue
- School of Pharmacy, Henan University, Kaifeng, China 475004
| | - Peilan Qi
- College of Medical Science, Henan Vocational University of Science and Technology, Zhoukou, China 466000.
| | - Mengke Zhang
- School of Pharmacy, Henan University, Kaifeng, China 475004
| | - Shiyong Song
- School of Pharmacy, Henan University, Kaifeng, China 475004.
| |
Collapse
|
11
|
Valente K, Boice GN, Polglase C, Belli RG, Bourque E, Suleman A, Brolo A. Synthesis of Gelatin Methacryloyl Analogs and Their Use in the Fabrication of pH-Responsive Microspheres. Pharmaceutics 2024; 16:1016. [PMID: 39204361 PMCID: PMC11360800 DOI: 10.3390/pharmaceutics16081016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024] Open
Abstract
pH-responsive hydrogels have numerous applications in tissue engineering, drug delivery systems, and diagnostics. Gelatin methacryloyl (GelMA) is a biocompatible, semi-synthetic polymer prepared from gelatin. When combined with aqueous solvents, GelMA forms hydrogels that have extensive applications in biomedical engineering. GelMA can be produced with different degrees of methacryloyl substitution; however, the synthesis of this polymer has not been tuned towards producing selectively modified materials for single-component pH-responsive hydrogels. In this work, we have explored two different synthetic routes targeting different gelatin functional groups (amine, hydroxyl, and/or carboxyl) to produce two GelMA analogs: gelatin A methacryloyl glycerylester (polymer A) and gelatin B methacrylamide (polymer B). Polymers A and B were used to fabricate pH-responsive hydrogel microspheres in a flow-focusing microfluidic device. At neutral pH, polymer A and B microspheres displayed an average diameter of ~40 µm. At pH 6, microspheres from polymer A showed a swelling ratio of 159.1 ± 11.5%, while at pH 10, a 288.6 ± 11.6% swelling ratio was recorded for polymer B particles.
Collapse
Affiliation(s)
- Karolina Valente
- VoxCell BioInnovation Inc., Victoria, BC V8T 5L2, Canada; (K.V.); (G.N.B.); (C.P.)
| | - Geneviève N. Boice
- VoxCell BioInnovation Inc., Victoria, BC V8T 5L2, Canada; (K.V.); (G.N.B.); (C.P.)
| | - Cameron Polglase
- VoxCell BioInnovation Inc., Victoria, BC V8T 5L2, Canada; (K.V.); (G.N.B.); (C.P.)
| | - Roman G. Belli
- Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada; (R.G.B.); (E.B.)
| | - Elaina Bourque
- Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada; (R.G.B.); (E.B.)
| | - Afzal Suleman
- Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada;
| | - Alexandre Brolo
- Department of Chemistry, University of Victoria, Victoria, BC V8P 5C2, Canada; (R.G.B.); (E.B.)
- Centre for Advanced Materials and Related Technology, University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
12
|
Long X, Liu M, Nan Y, Chen Q, Xiao Z, Xiang Y, Ying X, Sun J, Huang Q, Ai K. Revitalizing Ancient Mitochondria with Nano-Strategies: Mitochondria-Remedying Nanodrugs Concentrate on Disease Control. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308239. [PMID: 38224339 DOI: 10.1002/adma.202308239] [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: 08/14/2023] [Revised: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Mitochondria, widely known as the energy factories of eukaryotic cells, have a myriad of vital functions across diverse cellular processes. Dysfunctions within mitochondria serve as catalysts for various diseases, prompting widespread cellular demise. Mounting research on remedying damaged mitochondria indicates that mitochondria constitute a valuable target for therapeutic intervention against diseases. But the less clinical practice and lower recovery rate imply the limitation of traditional drugs, which need a further breakthrough. Nanotechnology has approached favorable regiospecific biodistribution and high efficacy by capitalizing on excellent nanomaterials and targeting drug delivery. Mitochondria-remedying nanodrugs have achieved ideal therapeutic effects. This review elucidates the significance of mitochondria in various cells and organs, while also compiling mortality data for related diseases. Correspondingly, nanodrug-mediate therapeutic strategies and applicable mitochondria-remedying nanodrugs in disease are detailed, with a full understanding of the roles of mitochondria dysfunction and the advantages of nanodrugs. In addition, the future challenges and directions are widely discussed. In conclusion, this review provides comprehensive insights into the design and development of mitochondria-remedying nanodrugs, aiming to help scientists who desire to extend their research fields and engage in this interdisciplinary subject.
Collapse
Affiliation(s)
- Xingyu Long
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, 750002, P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Yuting Xiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
| | - Jian Sun
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830017, P. R. China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, P. R. China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410078, P. R. China
| |
Collapse
|
13
|
Zhao Y, Jia C, Yao Z, Chen G, Huang G, Li H, Lu L, Jin T, Tang Y, Zhu Z, Zhang X. Dexamethasone Pretreatment Potentiates a Folic Acid-Functionalized Delivery System for Enhanced Lung Cancer Therapy. Mol Pharm 2024; 21:1077-1089. [PMID: 38346386 DOI: 10.1021/acs.molpharmaceut.3c00472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Folic acid (FA) has been widely engineered to promote the targeted delivery of FA-modified nanoparticles (NPs) by recognizing the folate receptor α (FRα). However, the efficacy of FA-targeted therapy significantly varied with the abundance of FRα and natural immunoglobulin levels in different tumors. Therefore, a sequential therapy of dexamethasone (Dex)-induced FRα amplification and immunosuppression combined with FA-functionalized doxorubicin (DOX) micelles to synergistically suppress tumor proliferation was proposed in this study. In brief, a pH/reduction-responsive FA-functionalized micelle (FCSD) was obtained by grafting FA, derivatization-modified cholesterol, and 2,3-dimethylmaleic anhydride onto a chitosan oligosaccharide. The obtained FCSD/DOX NPs can effectively deliver DOX in tumors, and their targeting efficiency can be further improved with Dex pretreatment to decrease the immunoglobulin M (IgM) content in serum and amplify FRα levels on the surface of M109 cells. After internalization, charge reversal and disulfide bond breakage of FCSD vectors under the stimulation of tumor extracellular pH (pHe) and intracellular glutathione (GSH) would contribute to the disintegration of vectors and the rapid release of DOX. The sequential therapy that combined Dex pretreatment and targeted chemotherapy by FCSD/DOX NPs demonstrated superior tumor suppression compared with monotherapy, which is expected to provide a potential strategy for FRα-positive lung cancer patients.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou 215003, China
| | - Changhao Jia
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Zhixin Yao
- School of Pharmacy, Yancheng Teachers' University, Yancheng 224002, China
| | - Gang Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Gui Huang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
- Children's Hospital of Wujiang District, Suzhou 215200, China
| | - Hui Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Linghong Lu
- Children's Hospital of Wujiang District, Suzhou 215200, China
| | - Taiwei Jin
- Children's Hospital of Wujiang District, Suzhou 215200, China
| | - Yan Tang
- School of Pharmacy, Yancheng Teachers' University, Yancheng 224002, China
| | - Zengyan Zhu
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou 215003, China
| | - Xuenong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| |
Collapse
|
14
|
Xu X, Ma J, Wang A, Zheng N. N-Sulfonyl amidine polypeptides: new polymeric biomaterials with conformation transition responsive to tumor acidity. Chem Sci 2024; 15:1769-1781. [PMID: 38303932 PMCID: PMC10829015 DOI: 10.1039/d3sc05504c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024] Open
Abstract
Manipulation of pH responsiveness is a frequently employed tactic in the formulation of trigger-responsive nanomaterials. It offers an avenue for "smart" designs capitalizing on distinctive pH gradients across diverse tissues and intracellular compartments. However, an overwhelming majority of documented functional groups (>80%) exhibit responsiveness solely to the heightened acidic milieu of intracellular pH (about 4.5-5.5). This scenario diverges markedly from the moderately acidic extracellular pH (∼6.8) characteristic of tumor microenvironments. Consequently, systems predicated upon intracellular pH responsiveness are unlikely to confer discernible advantages concerning targeted penetration and cellular uptake at tumor sites. In this study, we elucidated the extracellular pH responsiveness intrinsic to N-sulfonyl amidine (SAi), delineating a method to synthesize an array of SAi-bearing polypeptides (SAi-polypeptides). Notably, we demonstrated the pH-dependent modulation of SAi-polypeptide conformations, made possible by the protonation/deprotonation equilibrium of SAi in response to minute fluctuations in pH from physiological conditions to the extracellular milieu of tumors. This dynamic pH-triggered transition of SAi-polypeptides from negatively charged to neutrally charged side chains at the pH outside tumor cells (∼6.8) facilitated a transition from coil to helix conformations, concomitant with the induction of cellular internalization upon arrival at tumor sites. Furthermore, the progressive acidification of the intracellular environment expedited drug release, culminating in significantly enhanced site-specific chemotherapeutic efficacy compared with free-drug counterparts. The distinct pH-responsive attributes of SAi could aid the design of tumor acidity-responsive applications, thereby furnishing invaluable insights into the realm of smart material design.
Collapse
Affiliation(s)
- Xiang Xu
- School of Chemical Engineering, Dalian University of Technology Dalian 116024 China
| | - Jinjuan Ma
- Department of Comparative Medicine Laboratory Animal Center, Dalian Medical University Dalian 116000 China
| | - Aiguo Wang
- Department of Comparative Medicine Laboratory Animal Center, Dalian Medical University Dalian 116000 China
| | - Nan Zheng
- School of Chemical Engineering, Dalian University of Technology Dalian 116024 China
- Dalian University of Technology Corporation of Changshu Research Institution Suzhou 215500 China
| |
Collapse
|
15
|
Lyu M, Yazdi M, Lin Y, Höhn M, Lächelt U, Wagner E. Receptor-Targeted Dual pH-Triggered Intracellular Protein Transfer. ACS Biomater Sci Eng 2024; 10:99-114. [PMID: 35802884 DOI: 10.1021/acsbiomaterials.2c00476] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Protein therapeutics are of widespread interest due to their successful performance in the current pharmaceutical and medical fields, even though their broad applications have been hindered by the lack of an efficient intracellular delivery approach. Herein, we fabricated an active-targeted dual pH-responsive delivery system with favorable tumor cell entry augmented by extracellular pH-triggered charge reversal and tumor receptor targeting and pH-controlled endosomal release in a traceless fashion. As a traceable model protein, the enhanced green fluorescent protein (eGFP) bearing a nuclear localization signal was covalently coupled with a pH-labile traceless azidomethyl-methylmaleic anhydride (AzMMMan) linker followed by functionalization with different molar equivalents of two dibenzocyclooctyne-octa-arginine-cysteine (DBCO-R8C)-modified moieties: polyethylene glycol (PEG)-GE11 peptide for epidermal growth factor receptor-mediated targeting and melittin for endosomal escape. The cationic melittin domain was masked with tetrahydrophthalic anhydride revertible at mild acidic pH 6.5. At the optimally balanced ratio of functional units, the on-demand charge conversion at tumoral extracellular pH 6.5 in combination with GE11-mediated targeting triggered enhanced electrostatic cellular attraction by the R8C cell-penetrating peptides and melittin, as demonstrated by strongly enhanced cellular uptake. Successful endosomal release followed by nuclear localization of the eGFP cargo was obtained by taking advantage of melittin-mediated endosomal escape and rapid traceless release from the AzMMMan linker. The effectiveness of this multifunctional bioresponsive system suggests a promising strategy for delivery of protein drugs toward intracellular targets. A possible therapeutic relevance was indicated by an example of cytosolic delivery of cytochrome c initiating the apoptosis pathway to kill cancer cells.
Collapse
Affiliation(s)
- Meng Lyu
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Yi Lin
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Ulrich Lächelt
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, 1090 Vienna, Austria
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
16
|
Binaeian E, Nabipour H, Ahmadi S, Rohani S. The green synthesis and applications of biological metal-organic frameworks for targeted drug delivery and tumor treatments. J Mater Chem B 2023; 11:11426-11459. [PMID: 38047399 DOI: 10.1039/d3tb01959d] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Biological metal-organic frameworks (bio-MOFs) constitute a growing subclass of MOFs composed of metals and bio-ligands derived from biology, such as nucleobases, peptides, saccharides, and amino acids. Bio-ligands are more abundant than other traditional organic ligands, providing multiple coordination sites for MOFs. However, bio-MOFs are typically prepared using hazardous or harmful solvents or reagents, as well as laborious processes that do not conform to environmentally friendly standards. To improve biocompatibility and biosafety, eco-friendly synthesis and functionalization techniques should be employed with mild conditions and safer materials, aiming to reduce or avoid the use of toxic and hazardous chemical agents. Recently, bio-MOF applications have gained importance in some research areas, including imaging, tumor therapy, and targeted drug delivery, owing to their flexibility, low steric hindrances, low toxicity, remarkable biocompatibility, surface property refining, and degradability. This has led to an exponential increase in research on these materials. This paper provides a comprehensive review of updated strategies for the synthesis of environmentally friendly bio-MOFs, as well as an examination of the current progress and accomplishments in green-synthesized bio-MOFs for drug delivery aims and tumor treatments. In conclusion, we consider the challenges of applying bio-MOFs for biomedical applications and clarify the possible research orientation that can lead to highly efficient therapeutic outcomes.
Collapse
Affiliation(s)
- Ehsan Binaeian
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Hafezeh Nabipour
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Soroush Ahmadi
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada.
| | - Sohrab Rohani
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada.
| |
Collapse
|
17
|
Lee S, Lee SM, Lee SH, Choi WK, Park SJ, Kim DY, Oh SW, Oh J, Cho JY, Lee J, Chien PN, Nam SY, Heo CY, Lee YS, Kwak EA, Chung WJ. In situ photo-crosslinkable hyaluronic acid-based hydrogel embedded with GHK peptide nanofibers for bioactive wound healing. Acta Biomater 2023; 172:159-174. [PMID: 37832839 DOI: 10.1016/j.actbio.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023]
Abstract
A versatile hydrogel was developed for enhancing bioactive wound healing by introducing the amphiphilic GHK peptide (GHK-C16) into a photo-crosslinkable tyramine-modified hyaluronic acid (HA-Ty). GHK-C16 self-assembled into GHK nanofibers (GHK NF) in HA-Ty solution, which underwent in situ gelation after the wound area was filled with precursor solution. Blue light irradiation (460-490 nm), with riboflavin phosphate as a photoinitiator, was used to trigger crosslinking, which enhanced the stability of the highly degradable hyaluronic acid and enabled sustained release of the nanostructured GHK derivatives. The hydrogels provided a microenvironment that promoted the proliferation of dermal fibroblasts and the activation of cytokines, leading to reduced inflammation and increased collagen expression during wound healing. The complexation of Cu2+ into GHK nanofibers resulted in superior wound healing capabilities compared with non-lipidated GHK peptide with a comparable level of growth factor (EGF). Additionally, nanostructured Cu-GHK improved angiogenesis through vascular endothelial growth factor (VEGF) activation, which exerted a synergistic therapeutic effect. Furthermore, in vivo wound healing experiments revealed that the Cu-GHK NF/HA-Ty hydrogel accelerated wound healing through densely packed remodeled collagen in the dermis and promoting the growth of denser fibroblasts. HA-Ty hydrogels incorporating GHK NF also possessed improved mechanical properties and a faster wound healing rate, making them suitable for advanced bioactive wound healing applications. STATEMENT OF SIGNIFICANCE: By combining photo-crosslinkable tyramine-modified hyaluronic acid with self-assembled Cu-GHK-C16 peptide nanofibers (Cu-GHK NF), the Cu-GHK NF/HA-Ty hydrogel offers remarkable advantages over conventional non-structured Cu-GHK for wound healing. It enhances cell proliferation, migration, and collagen remodeling-critical factors in tissue regeneration. The incorporation of GHK nanofibers complexed with copper ions imparts potent anti-inflammatory effects, promoting cytokine activation and angiogenesis during wound healing. The Cu-GHK NF/hydrogel's unique properties, including in situ photo-crosslinking, ensure high customization and potency in tissue regeneration, providing a cost-effective alternative to growth factors. In vivo experiments further validate its efficacy, demonstrating significant wound closure, collagen remodeling, and increased fibroblast density. Overall, the Cu-GHK NF/HA-Ty hydrogel represents an advanced therapeutic option for wound healing applications.
Collapse
Affiliation(s)
- Seohui Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sang Min Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sang Hyun Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Woong-Ku Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sung-Jun Park
- School of Chemical and Biological Engineering, Seoul National University, 151-744, Seoul, Republic of Korea
| | - Do Yeon Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Sae Woong Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jieun Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Pham Ngoc Chien
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Sun Young Nam
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Chan Yeong Heo
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Medical Device Development, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yoon-Sik Lee
- School of Chemical and Biological Engineering, Seoul National University, 151-744, Seoul, Republic of Korea
| | - Eun-A Kwak
- Research Institute of Biomolecule Control, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea.
| | - Woo-Jae Chung
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea; Research Institute of Biomolecule Control, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do, 16419, Republic of Korea; Center for Biologics, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
18
|
Niu B, Wu Y, Zhou M, Lin R, Ge P, Chen X, Zhou H, Zhang X, Xie J. Precise delivery of celastrol by PEGylated aptamer dendrimer nanoconjugates for enormous therapeutic effect via superior intratumor penetration over antibody counterparts. Cancer Lett 2023; 579:216461. [PMID: 37898358 DOI: 10.1016/j.canlet.2023.216461] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023]
Abstract
Antibody-coated nanoparticles have been reported to have the extremely low delivery efficiency in solid tumors in preclinical trials. Though aptamers were considered to be superior over antibodies in cancer theranostics, whether PEGylated aptamer nanoparticles are better than antibody nanoparticles in improving delivery specificity and penetration efficiency of chemotherapeutics is still unknown. Here, we conjugate celastrol, a natural product with anti-tumor effect, onto PEGylated EpCAM aptamer or antibody dendrimers to obtain two nanoconjugates, and for the first time, conduct a comprehensive study to compare their performance in delivery specificity, intratumoral penetration ability and therapeutic outcomes. Our results showed that compared to antibody counterparts, PEGylated aptamer nanoconjugates exhibited the enhanced accumulation and retention specificities at tumor sites and the stronger intratumoral penetration capabilities by reducing the macrophage reservoir effects in solid tumors. When delivered celastrol to a colorectal xenograft tumor mice model by PEGylated aptamer dendrimers, 20 % of enhanced therapeutic efficiency was achieved compared to that by antibody-modified ones. Moreover, celastrol at 2 mg/kg delivered by PEGylated aptamer dendrimers showed the prominent anticancer efficiency (nearly 92 %) but without obvious side effects. These data firstly provide the proof-of-concept implementation that PEGylated aptamer nanoconjugates will display the great potential in the effective and safe cancer treatment with regard to the superiority over antibody ones in penetration abilities.
Collapse
Affiliation(s)
- Boning Niu
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China; Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuehuang Wu
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Min Zhou
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Ruimiao Lin
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Pengjin Ge
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Xiaohui Chen
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China.
| | - Xiaokun Zhang
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China
| | - Jingjing Xie
- School of Pharmaceutical Sciences, and Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiang'an South Road, Xiamen, Fujian, 361102, China.
| |
Collapse
|
19
|
Zhou Q, Xiang J, Qiu N, Wang Y, Piao Y, Shao S, Tang J, Zhou Z, Shen Y. Tumor Abnormality-Oriented Nanomedicine Design. Chem Rev 2023; 123:10920-10989. [PMID: 37713432 DOI: 10.1021/acs.chemrev.3c00062] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Anticancer nanomedicines have been proven effective in mitigating the side effects of chemotherapeutic drugs. However, challenges remain in augmenting their therapeutic efficacy. Nanomedicines responsive to the pathological abnormalities in the tumor microenvironment (TME) are expected to overcome the biological limitations of conventional nanomedicines, enhance the therapeutic efficacies, and further reduce the side effects. This Review aims to quantitate the various pathological abnormalities in the TME, which may serve as unique endogenous stimuli for the design of stimuli-responsive nanomedicines, and to provide a broad and objective perspective on the current understanding of stimuli-responsive nanomedicines for cancer treatment. We dissect the typical transport process and barriers of cancer drug delivery, highlight the key design principles of stimuli-responsive nanomedicines designed to tackle the series of barriers in the typical drug delivery process, and discuss the "all-into-one" and "one-for-all" strategies for integrating the needed properties for nanomedicines. Ultimately, we provide insight into the challenges and future perspectives toward the clinical translation of stimuli-responsive nanomedicines.
Collapse
Affiliation(s)
- Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Yechun Wang
- Department of Cell Biology, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- State Key Laboratory of Chemical Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
20
|
Lu Q, Yu H, Zhao T, Zhu G, Li X. Nanoparticles with transformable physicochemical properties for overcoming biological barriers. NANOSCALE 2023; 15:13202-13223. [PMID: 37526946 DOI: 10.1039/d3nr01332d] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
In recent years, tremendous progress has been made in the development of nanomedicines for advanced therapeutics, yet their unsatisfactory targeting ability hinders the further application of nanomedicines. Nanomaterials undergo a series of processes, from intravenous injection to precise delivery at target sites. Each process faces different or even contradictory requirements for nanoparticles to pass through biological barriers. To overcome biological barriers, researchers have been developing nanomedicines with transformable physicochemical properties in recent years. Physicochemical transformability enables nanomedicines to responsively switch their physicochemical properties, including size, shape, surface charge, etc., thus enabling them to cross a series of biological barriers and achieve maximum delivery efficiency. In this review, we summarize recent developments in nanomedicines with transformable physicochemical properties. First, the biological dilemmas faced by nanomedicines are analyzed. Furthermore, the design and synthesis of nanomaterials with transformable physicochemical properties in terms of size, charge, and shape are summarized. Other switchable physicochemical parameters such as mobility, roughness and mechanical properties, which have been sought after most recently, are also discussed. Finally, the prospects and challenges for nanomedicines with transformable physicochemical properties are highlighted.
Collapse
Affiliation(s)
- Qianqian Lu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| | - Hongyue Yu
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| | - Tiancong Zhao
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| | - Guanjia Zhu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, P. R. China.
| | - Xiaomin Li
- Department of Chemistry, Laboratory of Advanced Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials (2011-iChEM), Fudan University, Shanghai 200433, P. R. China.
| |
Collapse
|
21
|
Santra S, Das S, Sengupta A, Molla MR. Tumor acidity-induced surface charge modulation in covalent nanonetworks for activated cellular uptake: targeted delivery of anticancer drugs and selective cancer cell death. Biomater Sci 2023; 11:5549-5559. [PMID: 37401615 DOI: 10.1039/d3bm00491k] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023]
Abstract
A β-thioester and tertiary amine based covalently cross-linked nanoassembly coined as a nanonetwork (NN) endowed with dual pH responsive features (tumor acidity induced surface charge modulation and endosomal pH triggered controlled degradation) has been designed and synthesized for stable sequestration and sustained release of drug molecules in response to endosomal pH. An amphiphile integrated with tertiary amine and acrylate (ATA) functionalities was synthesized to fabricate the nanonetwork. This amphiphile showed entropically driven self-assembly and micellar nanostructures (nanoassemblies), which can sequester hydrophobic drug molecules at neutral pH. To further stabilize the nanoassemblies and the sequestered drug molecules even below its critical aggregation concentration (CAC), the micellar core was cross-linked via the thiol-acrylate Michael addition click reaction to generate multiple copies of acid labile β-thioester functionalities in the core, which undergo slow hydrolysis at endosomal pH (∼5.0), thus enabling sustained release of the anti-cancer drug doxorubicin at endosomal pH. The nanonetworks showed a significant decrease in drug leakage compared to the nanoassemblies (NAs), which was also justified by a low leakage coefficient calculated from the fluorescence resonance energy transfer experiment. The NN also exhibited dilution insensitivity and high serum stability, whereas the NA disassembled upon dilution and during serum treatment. The biological evaluation revealed tumor extracellular matrix pH (∼6.4-6.8) induced surface charge modulation and cancer cell (HeLa) selective activated cellular uptake of the doxorubicin loaded nanonetwork (NN-DOX). In contrast, the benign nature of NN-DOX towards normal cells (H9c2) suggests excellent cell specificity. Thus, we believe that the ease of synthesis, nanonetwork fabrication reproducibility, robust stability, smart nature of tumor microenvironment sensitive surface charge modulation, boosted tumoral-cell uptake, and triggered drug release will make this system a potential nanomedicine for chemotherapeutic treatments.
Collapse
Affiliation(s)
- Subrata Santra
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| | - Shreya Das
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata-700032, India
| | - Arunima Sengupta
- Department of Life Science & Biotechnology, Jadavpur University, Kolkata-700032, India
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| |
Collapse
|
22
|
Ding L, Zhang X, Yu P, Peng F, Sun Y, Wu Y, Luo Z, Li H, Zeng Y, Wu M, Liu X. Genetically engineered nanovesicles mobilize synergistic antitumor immunity by ADAR1 silence and PDL1 blockade. Mol Ther 2023; 31:2489-2506. [PMID: 37087570 PMCID: PMC10422002 DOI: 10.1016/j.ymthe.2023.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/04/2023] [Accepted: 04/18/2023] [Indexed: 04/24/2023] Open
Abstract
Growing evidence has proved that RNA editing enzyme ADAR1, responsible for detecting endogenous RNA species, was significantly associated with poor response or resistance to immune checkpoint blockade (ICB) therapy. Here, a genetically engineered nanovesicle (siAdar1-LNP@mPD1) was developed as an RNA interference nano-tool to overcome tumor resistance to ICB therapies. Small interfering RNA against ADAR1 (siAdar1) was packaged into a lipid nanoparticle (LNP), which was further coated with plasma membrane extracted from the genetically engineered cells overexpressing PD1. siAdar1-LNP@mPD1 could block the PD1/PDL1 immune inhibitory axis by presenting the PD1 protein on the coating membranes. Furthermore, siAdar1 could be effectively delivered into cancer cells by the designed nanovesicle to silence ADAR1 expression, resulting in an increased type I/II interferon (IFN-β/γ) production and making the cancer cells more sensitive to secreted effector cytokines such as IFN-γ with significant cell growth arrest. These integrated functions confer siAdar1-LNP@mPD1 with robust and comprehensive antitumor immunity, as evidenced by significant tumor growth regression, abscopal tumor prevention, and effective suppression of lung metastasis, through a global remodeling of the tumor immune microenvironment. Overall, we provided a promising translatable strategy to simultaneously silence ADAR1 and block PDL1 immune checkpoint to boost robust antitumor immunity.
Collapse
Affiliation(s)
- Lei Ding
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou 341000, P.R. China; School of Rare Earths, University of Science and Technology of China, Hefei 230026, P.R. China
| | - Xiaolong Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Peiwen Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Fang Peng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Yupeng Sun
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Yanni Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Zijin Luo
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Hongsheng Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Yongyi Zeng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China.
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China; The Liver Center of Fujian Province, Fujian Medical University, Fuzhou 350025, P.R. China; Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou 341000, P.R. China; School of Rare Earths, University of Science and Technology of China, Hefei 230026, P.R. China.
| |
Collapse
|
23
|
Wang X, Zhang H, Chen X, Wu C, Ding K, Sun G, Luo Y, Xiang D. Overcoming tumor microenvironment obstacles: Current approaches for boosting nanodrug delivery. Acta Biomater 2023; 166:42-68. [PMID: 37257574 DOI: 10.1016/j.actbio.2023.05.043] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/12/2023] [Accepted: 05/24/2023] [Indexed: 06/02/2023]
Abstract
In order to achieve targeted delivery of anticancer drugs, efficacy improvement, and side effect reduction, various types of nanoparticles are employed. However, their therapeutic effects are not ideal. This phenomenon is caused by tumor microenvironment abnormalities such as abnormal blood vessels, elevated interstitial fluid pressure, and dense extracellular matrix that affect nanoparticle penetration into the tumor's interstitium. Furthermore, nanoparticle properties including size, charge, and shape affect nanoparticle transport into tumors. This review comprehensively goes over the factors hindering nanoparticle penetration into tumors and describes methods for improving nanoparticle distribution by remodeling the tumor microenvironment and optimizing nanoparticle physicochemical properties. Finally, a critical analysis of future development of nanodrug delivery in oncology is further discussed. STATEMENT OF SIGNIFICANCE: This article reviews the factors that hinder the distribution of nanoparticles in tumors, and describes existing methods and approaches for improving the tumor accumulation from the aspects of remodeling the tumor microenvironment and optimizing the properties of nanoparticles. The description of the existing methods and approaches is followed by highlighting their advantages and disadvantages and put forward possible directions for the future researches. At last, the challenges of improving tumor accumulation in nanomedicines design were also discussed. This review will be of great interest to the broad readers who are committed to delivering nanomedicine for cancer treatment.
Collapse
Affiliation(s)
- Xiaohui Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Hong Zhang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, Shandong, 250033, China
| | - Xiaohui Chen
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Chunrong Wu
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Ke Ding
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China
| | - Guiyin Sun
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Debing Xiang
- Department of Oncology, Chongqing University Jiangjin Hospital, Chongqing 402260, China; Department of Oncology, Jiangjin Central Hospital of Chongqing, Chongqing 402260, China.
| |
Collapse
|
24
|
Kesharwani P, Prajapati SK, Jain A, Sharma S, Mody N, Jain A. Biodegradable Nanogels for Dermal Applications: An Insight. CURRENT NANOSCIENCE 2023; 19:509-524. [DOI: 10.2174/1573413718666220415095630] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/12/2021] [Accepted: 12/23/2021] [Indexed: 08/22/2024]
Abstract
Abstract:
Biodegradable nanogels in the biomedical field are emerging vehicles comprising
dispersions of hydrogel nanoparticles having 3D crosslinked polymeric networks. Nanogels show
distinguished characteristics including their homogeneity, adjustable size, low toxicity, stability
in serum, stimuli-responsiveness (pH, temperature, enzymes, light, etc.), and relatively good
drug encapsulation capability. Due to these characteristics, nanogels are referred to as nextgeneration
drug delivery systems and are suggested as promising carriers for dermal applications.
The site-specific delivery of drugs with effective therapeutic effects is crucial in transdermal drug
delivery. The nanogels made from biodegradable polymers can show external stimuliresponsiveness
which results in a change in gel volume, water content, colloidal stability, mechanical
strength, and other physical and chemical properties, thus improving the site-specific
topical drug delivery. This review provides insight into the advances in development, limitations,
and therapeutic significance of nanogels formulations. It also highlights the process of release of
drugs in response to external stimuli, various biodegradable polymers in the formulation of the
nanogels, and dermal applications of nanogels and their role in imaging, anti‐inflammatory therapy,
antifungal and antimicrobial therapy, anti‐psoriatic therapy, and ocular and protein/peptide
drug delivery.
Collapse
Affiliation(s)
- Payal Kesharwani
- Institute of Pharmacy, Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, 201310, Uttar Pradesh,
India
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O. Rajasthan 304022, India
| | - Shiv Kumar Prajapati
- Institute of Pharmacy, Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, 201310, Uttar Pradesh,
India
| | - Anushka Jain
- Raj Kumar
Goel Institute of Technology (Pharmacy), 5-Km. Stone, Delhi-Meerut Road, Ghaziabad, Uttar Pradesh, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, P.O. Rajasthan-304022-India
| | - Nishi Mody
- Department of Pharmaceutical Sciences, Dr. H. S. Gour University, Sagar (MP) 470003, India
| | - Ankit Jain
- Department of
Materials Engineering, Indian Institute of Science, Bangalore 560012 (Karnataka), India
| |
Collapse
|
25
|
Zhang K, Wang ZQ, Liu Z, Qu T, Zhang Z, Zeng F, Song H, Shi Q, Kang Z, Chen B, Dai P, Guo P, Tong Z, Xu W. Self-Disguised Nanospy for Improving Drug Delivery Efficiency via Decreasing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2300060. [PMID: 36929045 DOI: 10.1002/smll.202300060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Nanoscale drug carriers play a crucial role in reducing side effects of chemotherapy drugs. However, the mononuclear phagocyte system (MPS) and the drug protonation after nanoparticles (NPs) burst release still limit the drug delivery efficiency. In this work, a self-disguised Nanospy is designed to overcome this problem. The Nanospy is composed of: i) poly (lactic-co-glycolic acid)-polyethylene glycol (PLGA-PEG) loading doxorubicin is the core structure of the Nanospy. ii) CD47 mimic peptides (CD47p) is linked to NPs which conveyed the "don't eat me" signal. iii) 4-(2-aminoethyl) benzenesulfonamide (AEBS) as the inhibitor of Carbonic anhydrase IX (CAIX) linked to NPs. Briefly, when the Nanospy circulates in the bloodstream, CD47p binds to the regulatory protein α (SIRPα) on the surface of macrophages, which causes the Nanospy escapes from phagocytosis. Subsequently, the Nanospy enriches in tumor and the AEBS reverses the acidic microenvironment of tumor. Due to above characteristics, the Nanospy reduces liver macrophage phagocytosis by 25% and increases tumor in situ DOX concentration by 56% compared to PLGA@DOX treatment. In addition, the Nanospy effectively inhibits tumor growth with a 63% volume reduction. This work presents a unique design to evade the capture of MPS and overcomes the influence of acidic tumor microenvironment (TME) on weakly alkaline drugs.
Collapse
Affiliation(s)
- Kuo Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Zi-Qi Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Zhongqing Liu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Tianrui Qu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Zhishuai Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Fanshu Zeng
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Hongjian Song
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Qing Shi
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Zhijian Kang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Bogong Chen
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Peng Dai
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Pengyu Guo
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| | - Zhichao Tong
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, P. R. China
| | - Wanhai Xu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, P. R. China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin Medical University, Harbin, 150001, P. R. China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, P. R. China
| |
Collapse
|
26
|
Wang Q, Atluri K, Tiwari AK, Babu RJ. Exploring the Application of Micellar Drug Delivery Systems in Cancer Nanomedicine. Pharmaceuticals (Basel) 2023; 16:ph16030433. [PMID: 36986532 PMCID: PMC10052155 DOI: 10.3390/ph16030433] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Various formulations of polymeric micelles, tiny spherical structures made of polymeric materials, are currently being investigated in preclinical and clinical settings for their potential as nanomedicines. They target specific tissues and prolong circulation in the body, making them promising cancer treatment options. This review focuses on the different types of polymeric materials available to synthesize micelles, as well as the different ways that micelles can be tailored to be responsive to different stimuli. The selection of stimuli-sensitive polymers used in micelle preparation is based on the specific conditions found in the tumor microenvironment. Additionally, clinical trends in using micelles to treat cancer are presented, including what happens to micelles after they are administered. Finally, various cancer drug delivery applications involving micelles are discussed along with their regulatory aspects and future outlooks. As part of this discussion, we will examine current research and development in this field. The challenges and barriers they may have to overcome before they can be widely adopted in clinics will also be discussed.
Collapse
Affiliation(s)
- Qi Wang
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Keerthi Atluri
- Product Development Department, Alcami Corporation, Morrisville, NC 27560, USA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH 43614, USA
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Correspondence:
| |
Collapse
|
27
|
Yuan G, Liu Z, Wang W, Liu M, Xu Y, Hu W, Fan Y, Zhang X, Liu Y, Si G. Multifunctional nanoplatforms application in the transcatheter chemoembolization against hepatocellular carcinoma. J Nanobiotechnology 2023; 21:68. [PMID: 36849981 PMCID: PMC9969656 DOI: 10.1186/s12951-023-01820-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has the sixth-highest new incidence and fourth-highest mortality worldwide. Transarterial chemoembolization (TACE) is one of the primary treatment strategies for unresectable HCC. However, the therapeutic effect is still unsatisfactory due to the insufficient distribution of antineoplastic drugs in tumor tissues and the worsened post-embolization tumor microenvironment (TME, e.g., hypoxia and reduced pH). Recently, using nanomaterials as a drug delivery platform for TACE therapy of HCC has been a research hotspot. With the development of nanotechnology, multifunctional nanoplatforms have been developed to embolize the tumor vasculature, creating conditions for improving the distribution and bioavailability of drugs in tumor tissues. Currently, the researchers are focusing on functionalizing nanomaterials to achieve high drug loading efficacy, thorough vascular embolization, tumor targeting, controlled sustained release of drugs, and real-time imaging in the TACE process to facilitate precise embolization and enable therapeutic procedures follow-up imaging of tumor lesions. Herein, we summarized the recent advances and applications of functionalized nanomaterials based on TACE against HCC, believing that developing these functionalized nanoplatforms may be a promising approach for improving the TACE therapeutic effect of HCC.
Collapse
Affiliation(s)
- Gang Yuan
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Zhiyin Liu
- grid.488387.8Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Weiming Wang
- grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China ,grid.488387.8Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000 China
| | - Mengnan Liu
- grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China ,grid.488387.8National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yanneng Xu
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Wei Hu
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China ,grid.259384.10000 0000 8945 4455State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR China
| | - Yao Fan
- grid.410578.f0000 0001 1114 4286Department of Anus and Intestine Surgery, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China
| | - Xun Zhang
- grid.410578.f0000 0001 1114 4286Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000 China
| | - Yong Liu
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Guangyan Si
- Department of Intervention Radiology, Traditional Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
28
|
Kolarikova M, Hosikova B, Dilenko H, Barton-Tomankova K, Valkova L, Bajgar R, Malina L, Kolarova H. Photodynamic therapy: Innovative approaches for antibacterial and anticancer treatments. Med Res Rev 2023. [PMID: 36757198 DOI: 10.1002/med.21935] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 12/07/2022] [Accepted: 01/03/2023] [Indexed: 02/10/2023]
Abstract
Photodynamic therapy is an alternative treatment mainly for cancer but also for bacterial infections. This treatment dates back to 1900 when a German medical school graduate Oscar Raab found a photodynamic effect while doing research for his doctoral dissertation with Professor Hermann von Tappeiner. Unexpectedly, Raab revealed that the toxicity of acridine on paramecium depends on the intensity of light in his laboratory. Photodynamic therapy is therefore based on the administration of a photosensitizer with subsequent light irradiation within the absorption maxima of this substance followed by reactive oxygen species formation and finally cell death. Although this treatment is not a novelty, there is an endeavor for various modifications to the therapy. For example, selectivity and efficiency of the photosensitizer, as well as irradiation with various types of light sources are still being modified to improve final results of the photodynamic therapy. The main aim of this review is to summarize anticancer and antibacterial modifications, namely various compounds, approaches, and techniques, to enhance the effectiveness of photodynamic therapy.
Collapse
Affiliation(s)
- Marketa Kolarikova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Barbora Hosikova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hanna Dilenko
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Katerina Barton-Tomankova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Lucie Valkova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Robert Bajgar
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Lukas Malina
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Hana Kolarova
- Department of Biophysics, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
29
|
Bhaladhare S, Bhattacharjee S. Chemical, physical, and biological stimuli-responsive nanogels for biomedical applications (mechanisms, concepts, and advancements): A review. Int J Biol Macromol 2023; 226:535-553. [PMID: 36521697 DOI: 10.1016/j.ijbiomac.2022.12.076] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/30/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
The development of nanotechnology has influenced the advancements in biomedical and pharmaceutical fields. The design and formulation of stimuli-responsive nano-drug delivery systems, also called smart drug delivery systems, have attracted significant research worldwide and have been seen as a breakthrough in nanomedicines. The ability of these nanocarriers to respond to external and internal stimuli, such as pH, temperature, redox, electric and magnetic fields, enzymes, etc., has allowed them to deliver the cargo at targeted sites in a controlled fashion. The targeted drug delivery systems limit the harmful side effects on healthy tissue by toxic drugs and furnish spatial and temporal control drug delivery, improved patient compliance, and treatment efficiency. The polymeric nanogels (hydrogel nanoparticles) with stimuli-responsive characteristics have shown great potential in various biomedical, tissue engineering, and pharmaceutical fields. It is primarily because of their small size, biocompatibility, biodegradability, stimuli-triggered drug deliverability, high payload capacity, and tailored functionality. This comprehensive review deals distinctively with polymeric nanogels, their chemical, physical, and biological stimuli, the concepts of nanogels response to different stimuli, and recent advancements. This document will further improve the current understanding of stimuli-responsive materials and drug delivery systems and assist in exploring advanced potential applications of these intelligent materials.
Collapse
Affiliation(s)
- Sachin Bhaladhare
- Chemical and Polymer Engineering, Tripura University, Suryamaninagar, Tripura 799022, India.
| | - Sulagna Bhattacharjee
- Chemical and Polymer Engineering, Tripura University, Suryamaninagar, Tripura 799022, India
| |
Collapse
|
30
|
Khan RU, Shao J, Liao JY, Qian L. pH-triggered cancer-targeting polymers: From extracellular accumulation to intracellular release. NANO RESEARCH 2023; 16:5155-5168. [PMID: 36618069 PMCID: PMC9807988 DOI: 10.1007/s12274-022-5252-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 05/25/2023]
Abstract
Stimuli-responsive polymers are promising to achieve targeted delivery, improved stability during circulation, and controlled release of therapeutic and diagnostic agents. Among them, pH-responsive polymeric nanocarriers have attracted significant attention as pH varies in different body fluids (e.g., stomach, intestine, and colon) and intracellular organelles (e.g., endosome, lysosome, and mitochondria) to maintain homeostasis, while distinctive pH changes are also found in certain pathological states. For example, the extracellular environment of the tumor is acidic, which can be employed to drive selective delivery. During the internalization process, since most nanocarriers enter cells upon endocytosis where a drop of pH from 6.5 to 5.0 can occur from endosome to lysosome, pH-sensitive groups have been developed for enhanced cargo release. In this review, both non-covalent and covalent interactions responsive to pH changes are introduced, with a focus on the structure-property relationship and their applications in cancer targeting and endosomal escape.
Collapse
Affiliation(s)
- Rizwan Ullah Khan
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| | - Jinning Shao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| | - Jia-Yu Liao
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| | - Linghui Qian
- Institute of Drug Metabolism and Pharmaceutical Analysis, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Cancer Center & Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310058 China
| |
Collapse
|
31
|
Wang Y, Xia H, Chen B, Wang Y. Rethinking nanoparticulate polymer-drug conjugates for cancer theranostics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1828. [PMID: 35734967 DOI: 10.1002/wnan.1828] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 01/31/2023]
Abstract
Polymer-drug conjugates (PDCs) fabricated as nanoparticles have hogged the limelight in cancer theranostics in the past decade. Many researchers have devoted to developing novel and efficient polymeric drug delivery system since the first generation of poly(N-[2-hydroxypropyl]methacrylamide) copolymer-drug conjugates. However, none of them has been approved for chemotherapy in clinic. An ideal PDC nanoparticle for cancer theranostics should possess several properties, including prolonged circulation in blood, sufficient accumulation and internalization in tumors, and efficient drug release in target sites. To achieve these goals, it is important to rationally design the nanoparticulate PDCs based on circulation, accumulation, penetration, internalization, and drug release (CAPIR) cascade. Specifically, CAPIR cascades are divided into five steps: (1) circulation in the vascular compartment without burst release, (2) accumulation in tumors via enhanced permeability and retention effect, (3) subsequent penetration into the deep regions of tumors, (4) internalization into tumor cells, and (5) release of drugs as free molecules to exert their pharmacological effects. In this review, we focus on the development and novel approaches of nanoparticulate PDCs based on CAPIR cascade, and provide an outlook on future clinical application. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yaoqi Wang
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, China.,Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China.,Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
| | - Heming Xia
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
32
|
Wang C, Xu P, Li X, Zheng Y, Song Z. Research progress of stimulus-responsive antibacterial materials for bone infection. Front Bioeng Biotechnol 2022; 10:1069932. [PMID: 36636700 PMCID: PMC9831006 DOI: 10.3389/fbioe.2022.1069932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Infection is one of the most serious complications harmful to human health, which brings a huge burden to human health. Bone infection is one of the most common and serious complications of fracture and orthopaedic surgery. Antibacterial treatment is the premise of bone defect healing. Among all the antibacterial strategies, irritant antibacterial materials have unique advantages and the ability of targeted therapy. In this review, we focus on the research progress of irritating materials, the development of antibacterial materials and their advantages and disadvantages potential applications in bone infection.
Collapse
Affiliation(s)
| | | | | | - Yuhao Zheng
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| | - Zhiming Song
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
33
|
Lan T, Guo H, Lu X, Geng K, Wu L, Luo Y, Zhu J, Shen X, Guo Q, Wu S. Dual-Responsive Curcumin-Loaded Nanoparticles for the Treatment of Cisplatin-Induced Acute Kidney Injury. Biomacromolecules 2022; 23:5253-5266. [PMID: 36382792 DOI: 10.1021/acs.biomac.2c01083] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Acute kidney injury (AKI) has been a global public health concern leading to high patient morbidity and mortality in the world. Nanotechnology-mediated antioxidative therapy has facilitated the treatment of AKI. Herein, a hierarchical curcumin-loaded nanodrug delivery system (NPS@Cur) was fabricated for antioxidant therapy to ameliorate AKI. The nanoplatform could respond to subacidic and reactive oxygen species (ROS) microenvironments. The subacidic microenvironment led to a smaller size (from 140.9 to 99.36 nm) and positive charge (from -4.9 to 12.6 mV), contributing to the high accumulation of nanoparticles. An excessive ROS microenvironment led to nanoparticle degradation and drug release. In vitro assays showed that NPS@Cur could scavenge excessive ROS and relieve oxidative stress in H2O2-induced HK-2 cells through reduced apoptosis, activated autophagy, and decreased endoplasmic reticulum stress. Results from cisplatin-induced AKI models revealed that NPS@Cur could effectively alleviate mitochondria injury and protect kidneys via antioxidative protection, activated autophagy, decreased endoplasmic reticulum stress, and reduced apoptosis. NPS@Cur showed excellent biocompatibility and low toxicity to primary tissues in mice. These results revealed that NPS@Cur may be a potential therapeutic strategy for efficiently treating cisplatin or other cause-induced AKI.
Collapse
Affiliation(s)
- Tianyu Lan
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou510640, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou550025, China
| | - Honglei Guo
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing210029, China
| | - Xin Lu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou550025, China
| | - Kedui Geng
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou550025, China
| | - Lin Wu
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing210029, China
| | - Yongjun Luo
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou550025, China
| | - Jingfeng Zhu
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University (Jiangsu Province Hospital), Nanjing210029, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou550025, China
| | - Qianqian Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou550025, China
| | - Shuizhu Wu
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou510640, China
| |
Collapse
|
34
|
Design of Nanoparticles in Cancer Therapy Based on Tumor Microenvironment Properties. Pharmaceutics 2022; 14:pharmaceutics14122708. [PMID: 36559202 PMCID: PMC9785496 DOI: 10.3390/pharmaceutics14122708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, and battling cancer has always been a challenging subject in medical sciences. All over the world, scientists from different fields of study try to gain a deeper knowledge about the biology and roots of cancer and, consequently, provide better strategies to fight against it. During the past few decades, nanoparticles (NPs) have attracted much attention for the delivery of therapeutic and diagnostic agents with high efficiency and reduced side effects in cancer treatment. Targeted and stimuli-sensitive nanoparticles have been widely studied for cancer therapy in recent years, and many more studies are ongoing. This review aims to provide a broad view of different nanoparticle systems with characteristics that allow them to target diverse properties of the tumor microenvironment (TME) from nanoparticles that can be activated and release their cargo due to the specific characteristics of the TME (such as low pH, redox, and hypoxia) to nanoparticles that can target different cellular and molecular targets of the present cell and molecules in the TME.
Collapse
|
35
|
Developments on the Smart Hydrogel-Based Drug Delivery System for Oral Tumor Therapy. Gels 2022; 8:gels8110741. [PMID: 36421563 PMCID: PMC9689473 DOI: 10.3390/gels8110741] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022] Open
Abstract
At present, an oral tumor is usually treated by surgery combined with preoperative or postoperative radiotherapies and chemotherapies. However, traditional chemotherapies frequently result in substantial toxic side effects, including bone marrow suppression, malfunction of the liver and kidneys, and neurotoxicity. As a new local drug delivery system, the smart drug delivery system based on hydrogel can control drug release in time and space, and effectively alleviate or avoid these problems. Environmentally responsive hydrogels for smart drug delivery could be triggered by temperature, photoelectricity, enzyme, and pH. An overview of the most recent research on smart hydrogels and their controlled-release drug delivery systems for the treatment of oral cancer is given in this review. It is anticipated that the local drug release method and environment-responsive benefits of smart hydrogels will offer a novel technique for the low-toxicity and highly effective treatment of oral malignancy.
Collapse
|
36
|
Tuncaboylu DC, Wischke C. Opportunities and Challenges of Switchable Materials for Pharmaceutical Use. Pharmaceutics 2022; 14:2331. [PMID: 36365149 PMCID: PMC9696173 DOI: 10.3390/pharmaceutics14112331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 06/27/2024] Open
Abstract
Switchable polymeric materials, which can respond to triggering signals through changes in their properties, have become a major research focus for parenteral controlled delivery systems. They may enable externally induced drug release or delivery that is adaptive to in vivo stimuli. Despite the promise of new functionalities using switchable materials, several of these concepts may need to face challenges associated with clinical use. Accordingly, this review provides an overview of various types of switchable polymers responsive to different types of stimuli and addresses opportunities and challenges that may arise from their application in biomedicine.
Collapse
|
37
|
Zhang N, Feng N, Xin X, Zhang J, Wu D, Jiang Q, Yu T, Gao M, Zhao S, Yang H, Tian Q. Nano-drug delivery system with enhanced tumour penetration and layered anti-tumour efficacy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 45:102592. [PMID: 35905842 DOI: 10.1016/j.nano.2022.102592] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/31/2022] [Accepted: 07/20/2022] [Indexed: 06/15/2023]
Abstract
The low delivery efficiency of nano-drugs and limited tumour penetration are still huge challenges in treating solid tumours. Herein, we developed a pH-responsive nano-drug delivery system, CALS/PDMA@DOX, with a size conversion-layered delivery function. The system is composed of a pH-responsive cationic liposome loaded with DOX (CALS) and a polyamidoamine dendrimer loaded with DOX (PAMAM@DOX) modified with 2,3-dimethylmaleic anhydride (PDMA@DOX) using electrostatic adsorption. In the tumour microenvironment, the positively-charged large-size CALS and the positively-charged small-size PAMAM@DOX were dissociated to exert anti-tumour effects. CALS preferentially targeted tumour angiogenesis endothelial cells. Because of its small size and positive charge, PAMAM@DOX showed excellent tumour penetration. Significant tumour suppression by the system in vivo was confirmed in a 4T1 tumour xenograft mouse model. This pH-triggered size-switching layered delivery nanosystem is a safe and effective cancer treatment delivery platform that improves drug permeability and therapeutic efficacy.
Collapse
Affiliation(s)
- Nan Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Nannan Feng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiangying Xin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junwei Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Deqiao Wu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qianqian Jiang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Tong Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming Gao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Siyuan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hui Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qingfeng Tian
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
38
|
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: 45] [Impact Index Per Article: 15.0] [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
|
39
|
Li Z, Soroka IL, Tarakina NV, Sabatino MA, Muscolino E, Walo M, Jonsson M, Dispenza C. Inorganic/organic hybrid nanoparticles synthesized in a two-step radiation-driven process. Radiat Phys Chem Oxf Engl 1993 2022. [DOI: 10.1016/j.radphyschem.2022.110166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
40
|
Ren Z, Liao T, Li C, Kuang Y. Drug Delivery Systems with a "Tumor-Triggered" Targeting or Intracellular Drug Release Property Based on DePEGylation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5290. [PMID: 35955225 PMCID: PMC9369796 DOI: 10.3390/ma15155290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022]
Abstract
Coating nanosized anticancer drug delivery systems (DDSs) with poly(ethylene glycol) (PEG), the so-called PEGylation, has been proven an effective method to enhance hydrophilicity, aqueous dispersivity, and stability of DDSs. What is more, as PEG has the lowest level of protein absorption of any known polymer, PEGylation can reduce the clearance of DDSs by the mononuclear phagocyte system (MPS) and prolong their blood circulation time in vivo. However, the "stealthy" characteristic of PEG also diminishes the uptake of DDSs by cancer cells, which may reduce drug utilization. Therefore, dynamic protection strategies have been widely researched in the past years. Coating DDSs with PEG through dynamic covalent or noncovalent bonds that are stable in blood and normal tissues, but can be broken in the tumor microenvironment (TME), can achieve a DePEGylation-based "tumor-triggered" targeting or intracellular drug release, which can effectively improve the utilization of drugs and reduce their side effects. In this review, the stimuli and methods of "tumor-triggered" targeting or intracellular drug release, based on DePEGylation, are summarized. Additionally, the targeting and intracellular controlled release behaviors of the DDSs are briefly introduced.
Collapse
Affiliation(s)
- Zhe Ren
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China; (Z.R.); (T.L.)
| | - Tao Liao
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China; (Z.R.); (T.L.)
| | - Cao Li
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China; (Z.R.); (T.L.)
| | - Ying Kuang
- National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
41
|
Lu YF, Zhou JP, Zhou QM, Yang XY, Wang XJ, Yu JN, Zhang JG, Du YZ, Yu RS. Ultra-thin layered double hydroxide-mediated photothermal therapy combine with asynchronous blockade of PD-L1 and NR2F6 inhibit hepatocellular carcinoma. J Nanobiotechnology 2022; 20:351. [PMID: 35907841 PMCID: PMC9338598 DOI: 10.1186/s12951-022-01565-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/18/2022] [Indexed: 12/03/2022] Open
Abstract
Background The efficacy of immune checkpoint blockade (ICB), in the treatment of hepatocellular carcinoma (HCC), is limited due to low levels of tumor-infiltrating T lymphocytes and deficient checkpoint blockade in this immunologically "cool" tumor. Thus, combination approaches are needed to increase the response rates of ICB and induce synergistic antitumor immunity. Methods Herein, we designed a pH-sensitive multifunctional nanoplatform based on layered double hydroxides (LDHs) loaded with siRNA to block the intracellular immune checkpoint NR2F6, together with the asynchronous blockade surface receptor PD-L1 to induce strong synergistic antitumor immunity. Moreover, photothermal therapy (PTT) generated by LDHs after laser irradiation modified an immunologically “cold” microenvironment to potentiate Nr2f6-siRNA and anti-PD-L1 immunotherapy. Flow cytometry was performed to assess the immune responses initiated by the multifunctional nanoplatform. Results Under the slightly acidic tumor extracellular environment, PEG detached and the re-exposed positively charged LDHs enhanced tumor accumulation and cell uptake. The accumulated siRNA suppressed the signal of dual protumor activity in both immune and H22 tumor cells by silencing the NR2F6 gene, which further reduced the tumor burden and enhanced systemic antitumor immunity. The responses include enhanced tumor infiltration by CD4+ helper T cells, CD8+ cytotoxic T cells, and mature dendritic cells; the significantly decreased level of immune suppressed regulator T cells. The therapeutic responses were also attributed to the production of IL-2, IFN-γ, and TNF-α. The prepared nanoparticles also exhibited potential magnetic resonance imaging (MRI) ability, which could serve to guide synergistic immunotherapy treatment. Conclusions In summary, the three combinations of PTT, NR2F6 gene ablation and anti-PD-L1 can promote a synergistic immune response to inhibit the progression of primary HCC tumors and prevent metastasis. This study can be considered a proof-of-concept for the targeting of surface and intracellular immune checkpoints to supplement the existing HCC immunotherapy treatments. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01565-9.
Collapse
Affiliation(s)
- Yuan-Fei Lu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Jia-Ping Zhou
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Qiao-Mei Zhou
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Xiao-Yan Yang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Xiao-Jie Wang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Jie-Ni Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Jin-Guo Zhang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, People's Republic of China.
| | - Ri-Sheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People's Republic of China.
| |
Collapse
|
42
|
Zhang CX, Li HW, Zhang R, Ren Z, Wu Y. Tumor Microenvironments-Adaptive Apoptotic Effects of Cytidine 5'-monophosphate-Capped Gold Nanoclusters. ACS APPLIED BIO MATERIALS 2022; 5:3452-3460. [PMID: 35714365 DOI: 10.1021/acsabm.2c00380] [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: 11/29/2022]
Abstract
In the present work, cytidine 5'-monophosphate capped gold nanoclusters (AuNCs@CMP) are reported as a catalyst for redox reactions, which show both oxidase- and excellent peroxidase-like activity. When employing 3,3',5,5'-tetramethylbenzidine (TMB) as a substrate in the presence of hydrogen peroxide (H2O2), the maximum velocity (Vmax) was 175 × 10-8 M s-1 in vitro. Besides, the AuNCs@CMP exhibited high catalytic activity for reactive oxygen species (ROS) generation with H2O2. Particularly, they also displayed excellent catalytic activity for ROS generation in tumor cells, being activated and promoted by the tumor microenvironment (TME). Consequently, the AuNCs@CMP show an excellent antitumor effect on HeLa and SW480 cells as assayed by flow cytometry. The antitumor mechanism of AuNCs@CMP was attributed to the high ROS generation based on the specific environments of the TME. Therefore, the present study provides TME-adaptive AuNCs@CMP with excellent mimetic peroxidase activity, producing significant ROS to kill the tumor cells in TME.
Collapse
Affiliation(s)
- Chun-Xia Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Hong-Wei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| | - Renwen Zhang
- College of Chemical & Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Zhongyuan Ren
- College of Chemical & Pharmaceutical Engineering, Jilin Institute of Chemical Technology, Jilin 132022, P. R. China
| | - Yuqing Wu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, No. 2699 Qianjin Street, Changchun 130012, P. R. China.,Institute of Theoretical Chemistry, College of Chemistry, Jilin University, No. 2 Liutiao Road, Changchun 130023, P. R. China
| |
Collapse
|
43
|
Guo H, Liu F, Liu E, Wei S, Sun W, Liu B, Sun G, Lu L. Dual-responsive nano-prodrug micelles for MRI-guided tumor PDT and immune synergistic therapy. J Mater Chem B 2022; 10:4261-4273. [PMID: 35583206 DOI: 10.1039/d1tb02790e] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Micelles as nanocarriers not only offer new opportunities for early diagnosis and treatment of malignant cancers but also encounter numerous barriers in the path of efficient delivery of drugs to diseased areas in the body. To address these issues, we developed a pH/GSH responsive nano-prodrug micelle (NLG919/PGA-Cys-PPA@Gd) with a high drug-loading ratio and controlled drug release performance for MRI-guided tumor photodynamic therapy (PDT) and immune synergistic therapy. Under normal conditions, theranostic nanomicelles remained stable and in a photo-quenched state. Upon accumulation in the tumor site, however, the micelles demonstrated tumor microenvironment (TME) triggered photoactive formed-PPA (a photosensitizer) and NLG919 (an indoleamine 2,3-dioxygenase (IDO) inhibitor) release because the amide bonds of PGA-Cys-PPA and the disulfide linkage of Cys were sensitive to pH and GSH, respectively. More importantly, these micelles could avoid the undesired PPA leakage in blood circulation due to the conjugation between PPA and polymers. Furthermore, the obtained micelles could also enhance the contrast of T1-weighted MRI of tumors by virtue of their high relaxivity (r1 = 29.85 mM-1 s-1). In vitro and in vivo results illustrated that the micelles had good biocompatibility and biosafety. On the basis of the efficient drug delivery strategies in PDT and IDO pathway inhibition, this intelligent dual-drug delivery system could serve as an effective approach for MRI guided combination therapy of cancer.
Collapse
Affiliation(s)
- Hui Guo
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China. .,Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China
| | - Fangzhe Liu
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
| | - Enqi Liu
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China.
| | - Shanshan Wei
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China. .,Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China
| | - Wenbo Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China.
| | - Baoqiang Liu
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China. .,Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China
| | - Guoying Sun
- School of Chemistry and Life Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China. .,Advanced Institute of Materials Science, Changchun University of Technology, 2055 Yanan Street, Changchun 130012, P. R. China
| | - Lehui Lu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China.
| |
Collapse
|
44
|
A multi-hit therapeutic nanoplatform for hepatocellular carcinoma: Dual stimuli-responsive drug release, dual-modal imaging, and in situ oxygen supply to enhance synergistic therapy. Mater Today Bio 2022; 16:100338. [PMID: 35847375 PMCID: PMC9278082 DOI: 10.1016/j.mtbio.2022.100338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/07/2022] [Accepted: 06/18/2022] [Indexed: 12/26/2022]
Abstract
Nanomedicine has been widely studied for the diagnosis and treatment of hepatocellular carcinoma (HCC). How to synthesize a nanoplatform possessing a high synergistic therapeutic efficacy remains a challenge in this emerging research field. In this study, a convenient all-in-one therapeutic nanoplatform (FTY720@AM/T7-TL) is designed for HCC. This advanced nanoplatform consists of multiple functional elements, including gold-manganese dioxide nanoparticles (AM), tetraphenylethylene (T), fingolimod (FTY720), hybrid-liposome (L), and T7 peptides (T7). The nanoplatform is negatively charged at physiological pH and can transit to a positively charged state once moving to acidic pH environments. The specially designed pH-responsive charge-reversal nanocarrier prolongs the half-life of nanodrugs in blood and improves cellular uptake efficiency. The platform achieves a sustained and controllable drug release through dual stimulus-response, with pH as the endogenous stimulus and near-infrared as the exogenous stimulus. Furthermore, the nanoplatform realizes in situ O2 generation by catalyzing tumor over-expressed H2O2, which alleviates tumor microenvironment hypoxia and improves photodynamic therapy. Both in vitro and in vivo studies show the prepared nanoplatform has good photothermal conversion, cellular uptake efficiency, fluorescence/magnetic resonance imaging capabilities, and synergistic anti-tumor effects. These results suggest that the prepared all-in-one nanoplatform has great potential for dual-modal imaging-guided synergistic therapy of HCC.
Collapse
|
45
|
Abstract
Multicharged cyclodextrin (CD) supramolecular assemblies, including those based on positively/negatively charged modified mono-6-deoxy-CDs, per-6-deoxy-CDs, and random 2,3,6-deoxy-CDs, as well as parent CDs binding positively/negatively charged guests, have been extensively applied in chemistry, materials science, medicine, biological science, catalysis, and other fields. In this review, we primarily focus on summarizing the recent advances in positively/negatively charged CDs and parent CDs encapsulating positively/negatively charged guests, especially the construction process of supramolecular assemblies and their applications. Compared with uncharged CDs, multicharged CDs display remarkably high antiviral and antibacterial activity as well as efficient protein fibrosis inhibition. Meanwhile, charged CDs can interact with oppositely charged dyes, drugs, polymers, and biomacromolecules to achieve effective encapsulation and aggregation. Consequently, multicharged CD supramolecular assemblies show great advantages in improving drug-delivery efficiency, the luminescence properties of materials, molecular recognition and imaging, and the toughness of supramolecular hydrogels, in addition to enabling the construction of multistimuli-responsive assemblies. These features are anticipated to not only promote the development of CD-based supramolecular chemistry but also contribute to the rapid exploitation of these assemblies in diverse interdisciplinary applications.
Collapse
Affiliation(s)
- Zhixue Liu
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| | - Yu Liu
- State Key Laboratory of Elemento-Organic Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P. R. China. .,Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
46
|
Chen WH, Chen QW, Chen Q, Cui C, Duan S, Kang Y, Liu Y, Liu Y, Muhammad W, Shao S, Tang C, Wang J, Wang L, Xiong MH, Yin L, Zhang K, Zhang Z, Zhen X, Feng J, Gao C, Gu Z, He C, Ji J, Jiang X, Liu W, Liu Z, Peng H, Shen Y, Shi L, Sun X, Wang H, Wang J, Xiao H, Xu FJ, Zhong Z, Zhang XZ, Chen X. Biomedical polymers: synthesis, properties, and applications. Sci China Chem 2022; 65:1010-1075. [PMID: 35505924 PMCID: PMC9050484 DOI: 10.1007/s11426-022-1243-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 02/07/2023]
Abstract
Biomedical polymers have been extensively developed for promising applications in a lot of biomedical fields, such as therapeutic medicine delivery, disease detection and diagnosis, biosensing, regenerative medicine, and disease treatment. In this review, we summarize the most recent advances in the synthesis and application of biomedical polymers, and discuss the comprehensive understanding of their property-function relationship for corresponding biomedical applications. In particular, a few burgeoning bioactive polymers, such as peptide/biomembrane/microorganism/cell-based biomedical polymers, are also introduced and highlighted as the emerging biomaterials for cancer precision therapy. Furthermore, the foreseeable challenges and outlook of the development of more efficient, healthier and safer biomedical polymers are discussed. We wish this systemic and comprehensive review on highlighting frontier progress of biomedical polymers could inspire and promote new breakthrough in fundamental research and clinical translation.
Collapse
Affiliation(s)
- Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Chunyan Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350 China
| | - Shun Duan
- Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - Yongyuan Kang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Yun Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027 China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215 China
| | - Chengqiang Tang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Jinqiang Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Lei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Meng-Hua Xiong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006 China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123 China
| | - Kuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Zhanzhan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Xu Zhen
- Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093 China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Xiqun Jiang
- Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093 China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Huisheng Peng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027 China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215 China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Xuemei Sun
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006 China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190 China
| | - Fu-Jian Xu
- Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123 China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| |
Collapse
|
47
|
pH/ROS dual-responsive supramolecular polypeptide prodrug nanomedicine based on host-guest recognition for cancer therapy. Acta Biomater 2022; 143:381-391. [PMID: 35272024 DOI: 10.1016/j.actbio.2022.03.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/11/2022] [Accepted: 03/02/2022] [Indexed: 12/13/2022]
Abstract
Supramolecular nanomedicine assembly combined with polypeptide prodrug could become a powerful strategy to minimize drug leakage in blood circulation and trigger sufficient drug release at tumor tissue. Here, we developed a charge-reversal amphiphilic pillar[5]arene-modified polypeptide (P5-PLL-DMA), and reactive oxygen species (ROS)-sensitive polypeptide prodrug (P-PLL-DOX) including a ROS-cleavable thioketal (TK) linker between doxorubicin (DOX) and poly(L-lysine) (PLL), which could assemble via pillar[5]arene host-guest recognition, and further encapsulate chlorin e6 (Ce6) to obtain a supramolecular polypeptide prodrug (SPP-DOX/Ce6). The chemical conjugation to load drugs of DOX and the negatively charge of SPP-DOX/Ce6 could prevent premature drug leakage, and reduce undesirable interaction with serum proteins to enhance stability under physiological conditions (pH 7.4). Simultaneously, the carried charge of SPP-DOX/Ce6 reversed from negative to positive could effectively enhance the cellular internalization for efficient DOX delivery under acidic tumor microenvironment (pH 6.5). Upon 660 nm near-infrared light (NIR) irradiation, the ROS generated by encapsulated Ce6 rapidly cleaved the TK linker to release activated DOX, inducing the tumor-specific drug delivery. This intelligent supramolecular polypeptide prodrug based on pillar[5]arene host-guest recognition represents new avenues to develop stimulus responsive prodrug for enhanced cancer therapy with minimized the side effect. STATEMENT OF SIGNIFICANCE: In this work, a pH/ROS dual-sensitive supramolecular polypeptide prodrug (SPP-DOX/Ce6) was developed to minimize drug leakage in blood circulation and trigger sufficient drug release at tumor tissue. The chemical conjugation to load drugs of DOX via a ROS-cleavable thioketal (TK) linker and the distinctive charge-reversal capacity of SPP-DOX/Ce6 significantly enhances the stability under physiological conditions (pH 7.4), while facilitates cellular uptake at tumor site (pH 6.8). Upon 660 nm near-infrared light (NIR) irradiation, the ROS generated by encapsulated Ce6 induces the rapid cleavage of TK linker to release activated DOX, achieving a tumor-specific drug delivery. This intelligent supramolecular polypeptide prodrug SPP-DOX/Ce6 provides an effective strategy to construct stimulus responsive prodrug for enhanced cancer therapy.
Collapse
|
48
|
Cheng W, Su YL, Hsu HH, Lin YH, Chu LA, Huang WC, Lu YJ, Chiang CS, Hu SH. Rabies Virus Glycoprotein-Mediated Transportation and T Cell Infiltration to Brain Tumor by Magnetoelectric Gold Yarnballs. ACS NANO 2022; 16:4014-4027. [PMID: 35225594 DOI: 10.1021/acsnano.1c09601] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
T lymphocyte infiltration with immunotherapy potentially suppresses most devastating brain tumors. However, local immune privilege and tumor heterogeneity usually limit the penetration of immune cells and therapeutic agents into brain tumors, leading to tumor recurrence after treatment. Here, a rabies virus glycoprotein (RVG)-camouflaged gold yarnball (RVG@GY) that can boost the targeting efficiency at a brain tumor via dual hierarchy- and RVG-mediated spinal cord transportation, facilitating the decrease of tumor heterogeneity for T cell infiltration, is developed. Upon magnetoelectric irradiation, the electron current generated on the GYs activates the electrolytic penetration of palbociclib-loaded dendrimer (Den[Pb]) deep into tumors. In addition, the high-density GYs at brain tumors also induces the disruption of cell-cell interactions and T cell infiltration. The integration of the electrolytic effects and T cell infiltration promoted by drug-loaded RVG@GYs deep in the brain tumor elicits sufficient T cell numbers and effectively prolongs the survival rate of mice with orthotopic brain tumors.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei-Chen Huang
- Department of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| | - Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | | | | |
Collapse
|
49
|
Xie X, Zhang Y, Zhu Y, Lan Y. Preparation and Drug-Loading Properties of Amphoteric Cassava Starch Nanoparticles. NANOMATERIALS 2022; 12:nano12040598. [PMID: 35214927 PMCID: PMC8877468 DOI: 10.3390/nano12040598] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/01/2023]
Abstract
Based on the characteristics of charge reversal around the isoelectric point (pI) of amphoteric starch-containing anionic and cationic groups, amphoteric cassava starch nanoparticles (CA-CANPs) are prepared by a W/O microemulsion crosslinking method using (3-chloro-2-hydroxypropyl) trimethyl ammonium chloride as a cationic reagent and POCl3 as an anionic reagent, and the effects of preparation conditions on the particle size of the CA-CANPs are studied in detail in the present study. CA-CANPs with a smooth surface and an average diameter of 252 nm are successfully prepared at the following optimised conditions: a crosslinking agent amount of 15 wt%, an aqueous starch concentration of 6.0 wt%, an oil–water ratio of 10:1, a total surfactant amount of 0.20 g·mL−1, and a CHPTAC amount of 4.05 wt%. The pH-responsive value of the CA-CANPs can be regulated by adjusting the nitrogen–phosphorus molar ratio in the CA-CANPs. By using CA-CANPs with a pI of 6.89 as drug carriers and the paclitaxel (PTX) as a model drug, the maximum loading rate of 36.14 mg·g−1 is achieved, and the loading process is consistent with the Langmuir isotherm adsorption, with the calculated thermodynamic parameters of ΔH° = −37.91 kJ·mol−1, ΔS° = −10.96 J·mol−1·K−1 and ΔG° < 0. By testing the release rate in vitro, it is noted that the release rates of PTX in a neutral environment (37.6% after 96 h) and a slightly acidic environment (58.65% after 96 h) are quite different, suggesting that the CA-CANPs have the possibility of being a targeted controlled-release carrier with pH responsiveness for antitumor drugs.
Collapse
|
50
|
Zhang J, Lin W, Yang L, Zhang A, Zhang Y, Liu J, Liu J. Injectable and pH-responsive self-assembled peptide hydrogel for promoted tumor cell uptake and enhanced cancer chemotherapy. Biomater Sci 2022; 10:854-862. [PMID: 35006223 DOI: 10.1039/d1bm01788h] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chemotherapy is the main treatment for cancer therapy. However, its anti-tumor efficiency is always impaired by the poor bioavailability and low tumor accumulation of chemotherapeutic drugs. The variation between the tumor microenvironment and normal tissue has been recognized as an effective tool to improve drug anti-tumor efficiency. Herein, we developed an injectable, pH-responsive, in situ self-assembled drug-peptide hydrogel (MTX-KKFKFEFEF(DA)) for highly efficient local tumor chemotherapy with few side effects. The small molecule drug, methotrexate (MTX), and pH-responsive linker, 2,3-dimethylmaleic anhydride (DA), were facilely conjugated onto the chain of the KKFKFEFEF peptide via an amidation reaction. The negatively charged drug-peptide (pH 7.4) can be activated to be positive and achieve a sol-gel phase transition under an acidic microenvironment (pH 6.5) both in vitro and in vivo, resulting in highly efficient cellular uptake and endocytosis capacities. Moreover, the in vivo anti-tumor therapeutic effect revealed that the MTX-KKFKFEFEF(DA) hydrogel exhibits long-term tumor retention time, much better tumor inhibition rate and negligible side effects after intratumoral injection into breast tumor-bearing mice. Therefore, this study reveals a versatile strategy for fabricating a pH-responsive drug-peptide hydrogel to improve the chemotherapeutic efficacy of drugs in cancer treatment.
Collapse
Affiliation(s)
- Jiamin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Wenjing Lin
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Lijun Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Aijie Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Yumin Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, and Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| |
Collapse
|