1
|
Nsairat H, Lafi Z, Al-Najjar BO, Al-Samydai A, Saqallah FG, El-Tanani M, Oriquat GA, Sa’bi BM, Ibrahim AA, Dellinger AL, Alshaer W. How Advanced are Self-Assembled Nanomaterials for Targeted Drug Delivery? A Comprehensive Review of the Literature. Int J Nanomedicine 2025; 20:2133-2161. [PMID: 39990285 PMCID: PMC11847455 DOI: 10.2147/ijn.s490444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/22/2025] [Indexed: 02/25/2025] Open
Abstract
The development of effective drug delivery systems is a key focus in pharmaceutical research, aiming to enhance therapeutic efficacy while minimizing adverse effects. Self-assembled nanostructures present a promising solution due to their tunable properties, biocompatibility, and ability to encapsulate and deliver therapeutic agents to specific targets. This review examines recent advancements in drug-based self-assembled nanostructures for targeted delivery applications, including drug-drug conjugates, polymeric-based architectures, biomolecules, peptides, DNA, squalene conjugates and amphiphilic drugs. Various strategies for fabricating these nanostructures are discussed, with an emphasis on the design principles and mechanisms underlying their self-assembly and potential for targeted drug delivery to specific tissues or cells. Furthermore, the integration of targeting ligands, stimuli-responsive moieties and imaging agents into these nanostructures is explored for enhanced therapeutic outcomes and real-time monitoring. Challenges such as stability, scalability and regulatory hurdles in translating these nanostructures from bench to bedside are also addressed. Drug-based self-assembled nanostructures represent a promising platform for developing next-generation targeted drug delivery systems with improved therapeutic efficacy and reduced side effects.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Belal O Al-Najjar
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ali Al-Samydai
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Fadi G Saqallah
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Ghaleb Ali Oriquat
- Pharmacological and Diagnostic Research Center, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Bailasan Mohammad Sa’bi
- Pharmacological and Diagnostic Research Center, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Abed Alqader Ibrahim
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Anthony Lee Dellinger
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
2
|
Sayed Tabatabaei M, Sayed Tabatabaei FA, Moghimi HR. Drug self-delivery systems: A comprehensive review on small molecule nanodrugs. BIOIMPACTS : BI 2024; 15:30161. [PMID: 40161942 PMCID: PMC11954755 DOI: 10.34172/bi.30161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 04/02/2025]
Abstract
Drug self-delivery systems are nanostructures composed of a drug as the main structural unit, having the ability of intracellular trafficking with no additional carrier. In these systems, the drug itself undertakes the functional and structural roles; thereby, the ancillary role of excipients and carrier-related limitations are circumvented and therapeutic effect is achieved at a much lower dose. Such advantages -which are mainly but not exclusively beneficial in cancer treatment- have recently led to an upsurge of research on these systems. Subsequently, various terminologies were utilized to describe them, referring to the same concept with different words. However, not all the systems developed based on the self-delivery approach are introduced using one of these keywords. Using a scoping strategy, this review aims to encompass the systems that have been developed as yet -inspired by the concept of self-delivery- and classify them in a coherent taxonomy. Two main groups are introduced based on the type of building blocks: small molecule-based nanomedicines and self-assembling hybrid prodrugs. Due to the diversity, covering the whole gamut of topics is beyond the scope of a single article, and, inevitably, the latter is just briefly introduced here, whereas the features of the former group are meticulously presented. Depending on whether the drug is merely a carrier for itself or carries a second drug as cargo, two classes of small molecule-based nanomedicines are defined (i.e., pure nanodrugs and carrier-mimicking systems, respectively), each having sub-branches. After introducing each branch and giving some examples, possible strategies for designing each particular system are visually displayed. The resultant mind map can create a macro view of the taken path and its prospects, give a profound insight into opportunities, spark new ideas, and facilitate overcoming obstacles. Taken together, one can foresee a brilliant future for self-delivery systems as a pioneering candidate for the next generation of drug delivery systems.
Collapse
Affiliation(s)
- Mahsa Sayed Tabatabaei
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hamid Reza Moghimi
- Department of Pharmaceutics and Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Hu Y, Liu P. Design of pH/Redox Co-Triggered Degradable Diselenide-Containing Polyprodrug via a Facile One-Pot Two-Step Approach for Tumor-Specific Chemotherapy. Molecules 2024; 29:3837. [PMID: 39202916 PMCID: PMC11357291 DOI: 10.3390/molecules29163837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
The diselenide bond has attracted intense interest for drug delivery systems (DDSs) for tumor chemotherapy, owing to it possessing higher redox sensitivity than the disulfide one. Various redox-responsive diselenide-containing carriers have been developed for chemotherapeutics delivery. However, the premature drug leakage from these DDSs was significant enough to cause toxic side effects on normal cells. Here, a pH/redox co-triggered degradable polyprodrug was designed as a drug self-delivery system (DSDS) by incorporating drug molecules as structural units in the polymer main chains, using a facile one-pot two-step approach. The proposed PDOX could only degrade and release drugs by breaking both the neighboring acid-labile acylhydrazone and the redox-cleavable diselenide conjugations in the drug's structural units, triggered by the higher acidity and glutathione (GSH) or reactive oxygen species (ROS) levels in the tumor cells. Therefore, a slow solubility-controlled drug release was achieved for tumor-specific chemotherapy, indicating promising potential as a safe and efficient long-acting DSDS for future tumor treatment.
Collapse
Affiliation(s)
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
4
|
Guan Q, Liu Y, Xia Z, Zhang Y, Wang L, Wang Y, Zou S, Lv S, Zhou X. A novel nano-drug delivery system of glycyrrhetinic acid-mediated intracellular breakable brucine for enhanced anti-hepatitis B efficacy. J Biomater Appl 2024; 39:150-161. [PMID: 38748570 DOI: 10.1177/08853282241254750] [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: 06/23/2024]
Abstract
Background: Glycyrrhetinic acid-mediated brucine self-assembled nanomicelles enhance the anti-hepatitis B properties of brucine by improving its water solubility, short half-life, toxicity, and side effects. Brucine (B) is an indole alkaloid extracted from the seeds of Strychnos nux-vomica (Loganiaceae). Purpose: To assess the efficacy of the Brucine-Glycyrrhetnic acid-Polyethylene glycol-3,3'-dithiodipropionic acid-Glycerin monostearate (B-GPSG) in treating hepatitis B, its potential to protect against acute liver injury caused by d-galactosamine and its anti-hepatoma activities were studied. Research Design: The concentration of B-GPSG used in the in vivo and in vitro experiments was 0.63 mg/mL. The rats injected with d-GalN (450 mg/kg) were used as liver injury models. The rats were separated into normal, model, positive, positive control, B-PSG and B-GPSG groups. Hepatoma cells expressing HBV HepG2.2.15 were used for in vitro experiments. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, plate cloning, Hoechst staining and flow cytometry were conducted to explore the mechanism of B-GPSG against hepatitis B. Results: Compared with the model group, the liver coefficient of B-GPSG group decreased (4.59 ± 0.17 vs 5.88 ± 0.42), the content of MDA in rat liver homogenate decreased (12.54 ± 1.81 vs 23.05 ± 2.98), the activity of SOD increased, the activity of ALT and AST in rat serum decreased. In vitro, the IC50 values of B-GPSG group decreased. B-GPSG group effectively inhibited the proliferation and migration of HepG2.2.15 cells. Conclusions: The hepatoprotective effects of B-GPSG nanomicelles, which are attributed to their GA-mediated liver targeting and synergistic actions with brucine, suggest their therapeutic potential against hepatitis B. This development opens up new possibilities for the application of traditional Chinese medicine and nanomedicine in anti-hepatitis B.
Collapse
Affiliation(s)
- Qingxia Guan
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Yumeng Liu
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Zhaorui Xia
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Yue Zhang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Liping Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Yanhong Wang
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Shujun Zou
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Shaowa Lv
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| | - Xiaoying Zhou
- Key Laboratory of Basic and Application Research of Beiyao, Heilongjiang University of Chinese Medicine, Ministry of Education, 24 Heping Road, Xiangfang District, Harbin, Heilongjiang Province, China
| |
Collapse
|
5
|
Yang C, Liu P. Regulating Drug Release Performance of Acid-Triggered Dimeric Prodrug-Based Drug Self-Delivery System by Altering Its Aggregation Structure. Molecules 2024; 29:3619. [PMID: 39125024 PMCID: PMC11313937 DOI: 10.3390/molecules29153619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Dimeric prodrugs have been investigated intensely as carrier-free drug self-delivery systems (DSDSs) in recent decades, and their stimuli-responsive drug release has usually been controlled by the conjugations between the drug molecules, including the stimuli (pH or redox) and responsive sensitivity. Here, an acid-triggered dimeric prodrug of doxorubicin (DOX) was synthesized by conjugating two DOX molecules with an acid-labile ketal linker. It possessed high drug content near the pure drug, while the premature drug leakage in blood circulation was efficiently suppressed. Furthermore, its aggregation structures were controlled by fabricating nanomedicines via different approaches, such as fast precipitation and slow self-assembly, to regulate the drug release performance. Such findings are expected to enable better anti-tumor efficacy with the desired drug release rate, beyond the molecular structure of the dimeric prodrug.
Collapse
Affiliation(s)
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
6
|
Akpo E, Colin C, Perrin A, Cambedouzou J, Cornu D. Encapsulation of Active Substances in Natural Polymer Coatings. MATERIALS (BASEL, SWITZERLAND) 2024; 17:2774. [PMID: 38894037 PMCID: PMC11173946 DOI: 10.3390/ma17112774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Already used in the food, pharmaceutical, cosmetic, and agrochemical industries, encapsulation is a strategy used to protect active ingredients from external degradation factors and to control their release kinetics. Various encapsulation techniques have been studied, both to optimise the level of protection with respect to the nature of the aggressor and to favour a release mechanism between diffusion of the active compounds and degradation of the barrier material. Biopolymers are of particular interest as wall materials because of their biocompatibility, biodegradability, and non-toxicity. By forming a stable hydrogel around the drug, they provide a 'smart' barrier whose behaviour can change in response to environmental conditions. After a comprehensive description of the concept of encapsulation and the main technologies used to achieve encapsulation, including micro- and nano-gels, the mechanisms of controlled release of active compounds are presented. A panorama of natural polymers as wall materials is then presented, highlighting the main results associated with each polymer and attempting to identify the most cost-effective and suitable methods in terms of the encapsulated drug.
Collapse
Affiliation(s)
| | | | | | - Julien Cambedouzou
- IEM, Université de Montpellier, CNRS, ENSCM, F-34095 Montpellier, France
| | - David Cornu
- IEM, Université de Montpellier, CNRS, ENSCM, F-34095 Montpellier, France
| |
Collapse
|
7
|
Hu Y, Liu P. Diselenide-Bridged Doxorubicin Dimeric Prodrug: Synthesis and Redox-Triggered Drug Release. Molecules 2024; 29:1709. [PMID: 38675530 PMCID: PMC11052396 DOI: 10.3390/molecules29081709] [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: 02/14/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The diselenide bond has attracted intense interest in redox-responsive drug delivery systems (DDSs) in tumor chemotherapy, due to its higher sensitivity than the most investigated bond, namely the disulfide bond. Here, a diselenide-bridged doxorubicin dimeric prodrug (D-DOXSeSe) was designed by coupling two doxorubicin molecules with a diselenodiacetic acid (DSeDAA) molecule via α-amidation, as a redox-triggered drug self-delivery system (DSDS) for tumor-specific chemotherapy. The drug release profiles indicated that the D-DOXSeSe could be cleaved to release the derivatives selenol (DOX-SeH) and seleninic acid (DOX-SeOOH) with the triggering of high GSH and H2O2, respectively, indicating the double-edged sword effect of the lower electronegativity of the selenide atom. The resultant solubility-controlled slow drug release performance makes it a promising candidate as a long-acting DSDS in future tumor chemotherapy. Moreover, the interaction between the conjugations in the design of self-immolation traceless linkers was also proposed for the first time as another key factor for a desired precise tumor-specific chemotherapy, besides the conjugations themselves.
Collapse
Affiliation(s)
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
8
|
Xie P, Liu P. Formulation of DOX-dimer with bi-functionalized chitooligosaccharide for tumor-specific self-boosted drug release and synergistic chemo/chemodynamic therapy. Carbohydr Polym 2023; 320:121210. [PMID: 37659811 DOI: 10.1016/j.carbpol.2023.121210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/06/2023] [Accepted: 07/15/2023] [Indexed: 09/04/2023]
Abstract
The toxic side effects and possible drug resistance of the chemotherapeutics hinder their antitumor efficacy. Here, a pH/reactive oxygen species (ROS) dual-triggered nanodrug was developed for the tumor-specific self-boosted drug release and synergistic chemo/chemodynamic therapy, by formulating ROS-cleavable doxorubicin (DOX)-based dimer (DOX-TK-DOX) with bi-functionalized chitooligosaccharide (COS-Fc-TK) with ferrocenecarboxylic acid (Fc) and thioketal (TK). The resultant DOX-TK-DOX/COS-Fc-TK nanoparticles with a high DOX content of 39.70 % showed tumor-specific self-boosted drug release, which was triggered by highly toxic OH generated via Fc-catalyzed Fenton reaction of the endogenous H2O2 in tumor intracellular microenvironment. As a result, a synergistic chemo/chemodynamic therapy with combination index (CI) of 0.94 was achieved for selective treatment of tumors.
Collapse
Affiliation(s)
- Pengwei Xie
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
9
|
Liu P. Polyprodrugs for tumor chemotherapy: from molecular structure to drug release performance. J Mater Chem B 2023; 11:9565-9571. [PMID: 37791422 DOI: 10.1039/d3tb01700a] [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: 10/05/2023]
Abstract
Polyprodrugs have been recognized as promising carrier-free drug delivery systems (DDSs) for tumor chemotherapy in recent years, showing distinct superiority in comparison with the conventional polymer prodrugs. In the present work, the structure-property relationship of polyprodrugs was explored from the perspective of molecular structure, by discussing the effects of the conjugations and linkers on their drug content and drug releasing performance, including drug releasing rate and drug releasing selectivity, as well as the anti-tumor performance of the released drugs. Moreover, the future challenges in the design of polyprodrug-based DDSs with high anti-tumor efficacy were also highlighted.
Collapse
Affiliation(s)
- Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
10
|
Wei J, Liu Y, Li Y, Zhang Z, Meng J, Xie S, Li X. Photothermal Propelling and Pyroelectric Potential-Promoted Cell Internalization of Janus Nanoparticles and Pyroelectrodynamic Tumor Therapy. Adv Healthc Mater 2023; 12:e2300338. [PMID: 36857737 DOI: 10.1002/adhm.202300338] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Indexed: 03/03/2023]
Abstract
Cancer phototherapy experiences limitations in tissue diffusion and cell internalization of phototherapeutic agents and dose-dependent side effects. Herein, Janus pyroelectric nanoparticles (NPs) are designed to generate self-powered motion and built-in electric fields to overcome the delivery barriers. Polydopamine (PDA) layers are partially coated on tetragonal BaTiO3 (tBT) NPs to prepare Janus tBT@PDA, and Au NPs are deposited on the PDA caps to obtain Janus tBT@PDA-Au NPs. Near-infrared (NIR) illumination of tBT@PDA-Au builds in situ pyroelectric potentials on NPs, which selectively affect the membrane potential of tumor cells rather than normal cells to enhance tumor cell internalization and produce reactive oxygen species (ROS) for pyroelectric dynamic therapy (PEDT). The asymmetric photothermal effect of the Janus NPs creates thermophoretic force to propel NP motion, which enhances tumor diffusion and cellular uptake of NPs and boosts cytotoxicity and intracellular ROS levels. The inoculation of Au NPs increases the photothermal effect, exhibits larger motion velocities, produces higher pyroelectric potentials, and elevates cellular uptake rates, resulting in significant induction of tumor cell apoptosis, suppression of tumor growth, and extension of animal survival. Thus, the concise design of tBT@PDA-Au/NIR treatment has achieved thermophoretic motion-promoted tissue diffusion, built-in electric field-enhanced cell internalization, and photothermal/PEDT-synergized antitumor efficacy.
Collapse
Affiliation(s)
- Junwu Wei
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Yuan Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Yingxin Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Zhanlin Zhang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Jie Meng
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Shuang Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Xiaohong Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
11
|
Yu J, Xie X, Wang L, Liu W, Xu H, Lu X, Li X, Ren J, Li W. Smart Chondroitin Sulfate Micelles for Effective Targeted Delivery of Doxorubicin Against Breast Cancer Metastasis. Int J Nanomedicine 2023; 18:663-677. [PMID: 36798532 PMCID: PMC9926996 DOI: 10.2147/ijn.s398802] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Introduction Metastasis is a major challenge in breast cancer therapy. The successful chemotherapy of breast cancer largely depends on the ability to block the metastatic process. Herein, we designed a dual-targeting and stimuli-responsive drug delivery system for targeted drug delivery against breast cancer metastasis. Methods AS1411 aptamer-modified chondroitin sulfate A-ss-deoxycholic acid (ACSSD) was synthesized, and the unmodified CSSD was used as the control. Chemotherapeutic drug doxorubicin (DOX)-containing ACSSD (D-ACSSD) micelles were prepared by a dialysis method. The ACSSD conjugate was confirmed by Fourier transform infrared spectroscopy (FTIR), nuclear magnetic resonance (NMR), dynamic light scattering (DLS), and transmission electron microscopy (TEM). In vitro cellular uptake and cytotoxicity of D-ACSSD micelles were studied by confocal laser scanning microscopy (CLSM) and MTT assay in breast tumor cells. The inhibition capability of D-ACSSD micelles in cell migration and invasion was carried out in 4T1 cells. In vivo antitumor activity of DOX-containing micelles was investigated in metastatic 4T1-bearing Balb/c mice. Results D-ACSSD and DOX-loaded CSSD (D-CSSD) micelles exhibited high drug encapsulation content and reduction-responsive characteristics. D-ACSSD micelles were spherical in shape. Compared with D-CSSD, D-ACSSD showed higher cellular uptake and more potent killing activity in 4T1 and MDA-MB-231 cells. Additionally, D-ACSSD exhibited stronger inhibitory effects on the invasion and migration of highly metastatic 4T1 cells than unmodified D-CSSD. Among the DOX-containing formulations, D-ACSSD micelles presented the most effective inhibition of tumor growth and lung metastasis in orthotopic 4T1-bearing mice in vivo. It also revealed that ACSSD micelles did not exhibit obvious systemic toxicity. Conclusion The smart D-ACSSD micelles could be a promising delivery system for the therapy of metastatic breast cancer.
Collapse
Affiliation(s)
- Jingmou Yu
- Huzhou Key Laboratory of Medical and Environmental Applications Technologies, School of Life Sciences, Huzhou University, Huzhou, 313000, People’s Republic of China,Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Xin Xie
- Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Liangliang Wang
- Affiliated Hospital of Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Wenbo Liu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Huifeng Xu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Xiangmei Lu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Xiaofan Li
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China
| | - Jin Ren
- Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China,School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, 332000, People’s Republic of China,Correspondence: Jin Ren; Weidong Li, Email ;
| | - Weidong Li
- Jiangxi Provincial Key Laboratory of System Biomedicine, Jiujiang University, Jiujiang, 332000, People’s Republic of China,Jiujiang NO.1 People’s Hospital & Water of Life Hospital, Jiujiang, 332000, People’s Republic of China
| |
Collapse
|
12
|
Liu C, Liu C, Bai Y, Wang J, Tian W. Drug Self-Delivery Systems: Molecule Design, Construction Strategy, and Biological Application. Adv Healthc Mater 2022; 12:e2202769. [PMID: 36538727 DOI: 10.1002/adhm.202202769] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/29/2022] [Indexed: 02/01/2023]
Abstract
Drug self-delivery systems (DSDSs) offer new ways to create novel drug delivery systems (DDSs). In typical DSDSs, therapeutic reagents are not considered passive cargos but active delivery agents of actionable targets. As an advanced drug delivery strategy, DSDSs with positive cooperativity of both free drugs and nanocarriers exhibit the clear merits of unprecedented drug-loading capacity, minimized systemic toxicity, and flexible preparation of nanoscale deliverables for passive targeted therapy. This review highlights the recent advances and future trends in DSDSs on the basis of two differently constructed structures: covalent and noncovalent bond-based DSDSs. Specifically, various chemical and architectural designs, fabrication strategies, and responsive and functional features are comprehensively discussed for these two types of DSDSs. In addition, additional comments on the current development status of DSDSs and the potential applications of their molecular designs are presented in the corresponding discussion. Finally, the promising potential of DSDSs in biological applications is revealed and the relationship between preliminary molecular design of DSDSs and therapeutic effects of subsequent DSDSs biological applications is clarified.
Collapse
Affiliation(s)
- Chengfei Liu
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Caiping Liu
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, China
| | - Yang Bai
- Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science and Technology, Xi'an, Shaanxi, 710021, China
| | - Jingxia Wang
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Wei Tian
- Shaanxi Key Laboratory of Macromolecular Science and Technology, MOE Key Laboratory of Material Physics and Chemistry under Extraordinary Conditions, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| |
Collapse
|
13
|
Wei L, Zhang D, Xiong B, Zhang S, Zu Y, Jiang S. Inhibition of metastatic bone cancer with a cascade targeting of docetaxel. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
14
|
Ren G, Duan D, Wang G, Wang R, Li Y, Zuo H, Zhang Q, Zhang G, Zhao Y, Wang R, Zhang S. Construction of reduction-sensitive heterodimer prodrugs of doxorubicin and dihydroartemisinin self-assembled nanoparticles with antitumor activity. Colloids Surf B Biointerfaces 2022; 217:112614. [PMID: 35700564 DOI: 10.1016/j.colsurfb.2022.112614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 11/20/2022]
Abstract
Doxorubicin (DOX) is used as a first-line chemotherapeutic drug, whereas dihydroartemisinin (DHA) also shows a certain degree of antitumor activity. Disulfide bonds (-SS-) in prodrug molecules can be degraded in highly reducing environments. Thus, heterodimer prodrugs of DOX and DHA linked by a disulfide bond was designed and subsequently prepared as reduction-responsive self-assembled nanoparticles (DOX-SS-DHA NPs). In an in vitro release study, DOX-SS-DHA NPs exhibited reduction-responsive activity. Upon cellular evaluation, DOX-SS-DHA NPs were found to have better selectivity toward tumor cells and less cytotoxicity to normal cells. Compared to free DiR, DOX-SS-DHA NPs showed improved accumulation at the tumor site and even had a longer clearance half-life. More importantly, DOX-SS-DHA NPs possessed a much higher tumor inhibition efficacy than DOX-sol and MIX-sol in 4T1 tumor-bearing mice. Our results suggested the superior antitumor efficacy of DOX-SS-DHA NPs with less cytotoxicity.
Collapse
Affiliation(s)
- Guolian Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Danyu Duan
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Geng Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rongrong Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yujie Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hengtong Zuo
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qichao Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guoshun Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongdan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruili Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
15
|
Fu Y, Bian X, Li P, Huang Y, Li C. Carrier-Free Nanomedicine for Cancer Immunotherapy. J Biomed Nanotechnol 2022; 18:939-956. [PMID: 35854464 DOI: 10.1166/jbn.2022.3315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
With the rapid development of nanotechnology, carrier-based nano-drug delivery systems (DDSs) have been widely studied due to their advantages in optimizing pharmacokinetic and distribution profiles. However, despite those merits, some carrier-related limitations, such as low drug-loading capacity, systematic toxicity and unclear metabolism, usually prevent their further clinical transformation. Carrier-free nanomedicines with non-therapeutic excipients, are considered as an excellent paradigm to overcome these obstacles, owing to their superiority in improving both drug delivery efficacy and safety concern. In recent years, carrier-free nanomedicines have opened new horizons for cancer immunotherapy, and have already made outstanding progress. Herein, in this review, we are focusing on making an integrated and exhaustive overview of lately reports about them. Firstly, the major synthetic strategies of carrier-free nanomedicines are introduced, such as nanocrystals, prodrug-, amphiphilic drug-drug conjugates (ADDCs)-, polymer-drug conjugates-, and peptide-drug conjugates (PepDCs)-assembled nanomedicines. Afterwards, the typical applications of carrier-free nanomedicines in cancer immunotherapy are well-discussed, including cancer vaccines, cytokine therapy, enhancing T-cell checkpoint inhibition, as well as modulating tumor microenvironment (TME). After that, both the advantages and the potential challenges, as well as the future prospects of carrier-free nanomedicines in cancer immunotherapy, were discussed. And we believe that it would be of great potential practiced and reference value to the relative fields.
Collapse
Affiliation(s)
- Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Pingrong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| |
Collapse
|
16
|
He S, Meng Q, Zhong S, Gao Y, Cui X. Sonochemical fabrication of reduction-responsive alginate-based nanocapsules with folate targeting for drug delivery. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.128349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
17
|
Li J, Li X, Xie P, Liu P. Regulation of drug release performance using mixed doxorubicin-doxorubicin dimer nanoparticles as a pH-triggered drug self-delivery system. J Pharm Anal 2022; 12:122-128. [PMID: 35573875 PMCID: PMC9073254 DOI: 10.1016/j.jpha.2021.03.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 02/25/2021] [Accepted: 03/05/2021] [Indexed: 11/08/2022] Open
Abstract
A mixed drug self-delivery system (DSDS) with high drug content (>50%) was developed to regulate pH-triggered drug release, based on two doxorubicin (DOX)-DOX dimmers: D-DOXADH and D-DOXcar conjugated with acid-labile dynamic covalent bonds (hydrazone and carbamate, respectively) and stabilized with PEGylated D-DOXADH (D-DOXADH-PEG). Owing to the different stability of the dynamic covalent bonds in the two dimers and the noncovalent interaction between them, pH-triggered drug release could be easily regulated by adjusting the feeding ratios of the two DOX-DOX dimers in the mixed DSDS. Similar in vitro cellular toxicity was achieved with the mixed DSDS nanoparticles prepared with different feeding ratios. The mixed DSDS nanoparticles had a similar DOX content and diameter but different drug releasing rates. The MTT assays revealed that a high anti-tumor efficacy could be achieved with the slow-release mixed DSDS nanoparticles.
Collapse
Affiliation(s)
- Jiagen Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Pengwei Xie
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
18
|
Souri M, Soltani M, Moradi Kashkooli F, Kiani Shahvandi M, Chiani M, Shariati FS, Mehrabi MR, Munn LL. Towards principled design of cancer nanomedicine to accelerate clinical translation. Mater Today Bio 2022; 13:100208. [PMID: 35198957 PMCID: PMC8841842 DOI: 10.1016/j.mtbio.2022.100208] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/08/2023] Open
Abstract
Nanotechnology in medical applications, especially in oncology as drug delivery systems, has recently shown promising results. However, although these advances have been promising in the pre-clinical stages, the clinical translation of this technology is challenging. To create drug delivery systems with increased treatment efficacy for clinical translation, the physicochemical characteristics of nanoparticles such as size, shape, elasticity (flexibility/rigidity), surface chemistry, and surface charge can be specified to optimize efficiency for a given application. Consequently, interdisciplinary researchers have focused on producing biocompatible materials, production technologies, or new formulations for efficient loading, and high stability. The effects of design parameters can be studied in vitro, in vivo, or using computational models, with the goal of understanding how they affect nanoparticle biophysics and their interactions with cells. The present review summarizes the advances and technologies in the production and design of cancer nanomedicines to achieve clinical translation and commercialization. We also highlight existing challenges and opportunities in the field.
Collapse
Key Words
- CFL, Cell-free layer
- CGMD, Coarse-grained molecular dynamic
- Clinical translation
- DPD, Dissipative particle dynamic
- Drug delivery
- Drug loading
- ECM, Extracellular matrix
- EPR, Permeability and retention
- IFP, Interstitial fluid pressure
- MD, Molecular dynamic
- MDR, Multidrug resistance
- MEC, Minimum effective concentration
- MMPs, Matrix metalloproteinases
- MPS, Mononuclear phagocyte system
- MTA, Multi-tadpole assemblies
- MTC, Minimum toxic concentration
- Nanomedicine
- Nanoparticle design
- RBC, Red blood cell
- TAF, Tumor-associated fibroblast
- TAM, Tumor-associated macrophage
- TIMPs, Tissue inhibitor of metalloproteinases
- TME, Tumor microenvironment
- Tumor microenvironment
Collapse
Affiliation(s)
- Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran
| | | | | | - Mohsen Chiani
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Lance L. Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
19
|
Ma J, Chen Y, Liang W, Li L, Du J, Pan C, Zhang C. ROS-responsive dimeric prodrug-based nanomedicine targeted therapy for gastric cancer. Drug Deliv 2021; 28:1204-1213. [PMID: 34142633 PMCID: PMC8218932 DOI: 10.1080/10717544.2021.1937380] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) remains a major public health problem. Ursolic acid (UA) is reported to be effective in inhibiting GC; however, its low solubility and poor biocompatibility have greatly hindered its clinical application. Herein, an innovative reactive oxygen species (ROS)-sensitive UA dimeric prodrug is developed by coupling two UA molecules via a ROS-cleavable linkage, which can self-assemble into stable nanoparticles in the presence of surfactant. This new UA-based delivery system comprises the following major components: (I) dimeric prodrug inner core that can achieve high drug-loading (55%, w/w) and undergo rapid and selective conversion into intact drug molecules in response to ROS; (II) a polyethylene glycol (PEG) shell to improve colloid stability and extend blood circulation, and (III) surface-modified internalizing RGD (iRGD) to increase tumor targeting. Enhancement of the antitumor effect of this delivery system was demonstrated against GC tumors in vitro and in vivo. This novel approach offers the potential for clinical applications of UA.
Collapse
Affiliation(s)
- Jiachi Ma
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Yuzhong Chen
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Wanqing Liang
- Bengbu Medical College, Bengbu, People's Republic of China
| | - Lei Li
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Jun Du
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Chengwu Pan
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| | - Chensong Zhang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, People's Republic of China
| |
Collapse
|
20
|
Li G, Sun B, Li Y, Luo C, He Z, Sun J. Small-Molecule Prodrug Nanoassemblies: An Emerging Nanoplatform for Anticancer Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101460. [PMID: 34342126 DOI: 10.1002/smll.202101460] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/21/2021] [Indexed: 06/13/2023]
Abstract
The antitumor efficiency and clinical translation of traditional nanomedicines is mainly restricted by low drug loading, complex preparation technology, and potential toxicity caused by the overused carrier materials. In recent decades, small-molecule prodrug nanoassemblies (SMP-NAs), which are formed by the self-assembly of prodrugs themselves, have been widely investigated with distinct advantages of ultrahigh drug-loading and negligible excipients-trigged adverse reaction. Benefited from the simple preparation process, SMP-NAs are widely used for chemotherapy, phototherapy, immunotherapy, and tumor diagnosis. In addition, combination therapy based on the accurate co-delivery behavior of SMP-NAs can effectively address the challenges of tumor heterogeneity and multidrug resistance. Recent trends in SMP-NAs are outlined, and the corresponding self-assembly mechanisms are discussed in detail. Besides, the smart stimuli-responsive SMP-NAs and the combination therapy based on SMP-NAs are summarized, with special emphasis on the structure-function relationships. Finally, the outlooks and potential challenges of SMP-NAs in cancer therapy are highlighted.
Collapse
Affiliation(s)
- Guanting Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yaqiao Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
21
|
Li J, Li X, Liu P. Acid-triggered degradable diblock poly(doxorubicin)-polyethylene glycol polyprodrug with doxorubicin as structural unit for tumor intracellular delivery. Int J Pharm 2021; 609:121142. [PMID: 34600057 DOI: 10.1016/j.ijpharm.2021.121142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/30/2021] [Accepted: 09/25/2021] [Indexed: 10/20/2022]
Abstract
Polyprodrugs, in which drug was used as the structural unit by linking with each other via the dynamic covalent bonds in the main chain, are expected to endow excellent drug delivery performance. Here, acid-triggered degradable diblock polyprodrug, poly(doxorubicin)-polyethylene glycol (PDOX-PEG), was designed with DOX as structural unit alternately linked with acid-labile hydrazone and maleic amide groups, by the polycondensation of DOX-based dimers (D-DOXADH or D-DOXMAH) with PEGylated dimer (DOX-ADH-DOX-PEG) as end capping agent. The optimized PDOX-PEG, which was synthesized with D-DOXADH and the PEGylated dimer at a feeding ratio of 10%, possessed a high Mn of 3.1 × 104 g/mol with a high DOX content of 75.42%. It could easily self-assemble into near spherical nanoparticles with average hydrodynamic diameter of 135 nm. They showed excellent pH-triggered sustained drug release owing to the acid-triggered degradation of the polyprodrug block in the tumor intracellular microenvironment, with low premature drug leakage of 4.39 % within 60 h. The MTT results indicated the enhanced antitumor efficacy of the proposed PDOX-PEG nanoparticles than free DOX. The results demonstrated the promising potential of the proposed acid-triggered degradable diblock PDOX-PEG polyprodrug for tumor treatment.
Collapse
Affiliation(s)
- Jiagen Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
22
|
Gu G, Chen C, Zhang S, Yin B, Wang J. Self-Assembly Dual-Responsive NO Donor Nanoparticles for Effective Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50682-50694. [PMID: 34668695 DOI: 10.1021/acsami.1c12646] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Drug resistance and the serious side effects caused by classical chemotherapy drugs necessitate the development of novel targeted drug delivery systems. The high lipophilicity and short half-life of nitric oxide (NO), a gas with strong antitumor activity, make it difficult to reach the tumor site and result in a poor therapeutic effect in vivo. In order to overcome the deficiencies of the existing NO donors and NO delivery vehicles, a novel strategy was proposed to deliver NO for cancer chemotherapy by the prodrug dimer self-assembly nanoparticles of NO donors. Specifically, phenylsulfonylfuroxan (FZ) was chosen as the NO donor to synthesize the prodrug dimer precursor (FZ-SS-FZ) by disulfide linkages and ester bonds. The insertion of disulfide linkages promotes the self-assembly of FZ-SS-FZ in water. After this, the dual-responsive and tumor-targeting NO delivery system (FZ-SS-FZ@FA NPs) will finally be fabricated by further introducing folic acid on the surface of nanoparticles. FZ-SS-FZ can self-assemble to form uniform nanoparticles in water, which can effectively deliver NO to the tumor site and be uptaken by tumor cells, thus resulting in specific NO release in tumor cells and inducing tumor cell apoptosis. FZ-SS-FZ@FA NPs significantly improve the drug loading and delivery efficiencies of NO for chemotherapy, while enhancing its efficacy, providing a novel strategy for the tumor-targeted delivery of NO and at the same time laying a theoretical basis for the clinical translation of NO-based gas chemotherapy, opening up a new approach for cancer chemotherapy.
Collapse
Affiliation(s)
- Guolong Gu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, People's Republic of China
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Chen Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, People's Republic of China
| | - Shichao Zhang
- School of Pharmacy, Yancheng Teachers University, Yancheng 224007, People's Republic of China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, People's Republic of China
- Institutes of Integrative Medicine, Fudan University, Shanghai 201203, People's Republic of China
| |
Collapse
|
23
|
Davidovic T, Schimpf J, Sprenger-Mähr H, Abbassi-Nik A, Soleiman A, Zitt E, Lhotta K. Preparation and evaluation of reduction-responsive micelles based on disulfide-linked chondroitin sulfate A-tocopherol succinate for controlled antitumour drug release. J Pharm Pharmacol 2021; 73:1405-1417. [PMID: 34254648 PMCID: PMC8556126 DOI: 10.1093/jpp/rgab096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/10/2021] [Indexed: 11/14/2022]
Abstract
OBJECTIVES The study was to construct reduction-responsive chondroitin sulfate A (CSA)-conjugated TOS (CST) micelles with disulfide bond linkage, which was used for controlled doxorubicin (DOX) release and improved drug efficacy in vivo. METHODS CST and non-responsive CSA-conjugated TOS (CAT) were synthesized, and the chemical structure was confirmed by Fourier transform infrared (FTIR) spectroscopy, proton nuclear magnetic resonance (1H NMR) spectroscopy, fluorescence spectrophotometer and dynamic light scattering. Antitumour drug DOX was physically encapsulated into CST and CSA by dialysis method. Cell uptake of DOX-based formulations was investigated by confocal laser scanning microscopy. In vitro cytotoxicity was studied in A549 and AGS cells. Furthermore, antitumour activity was evaluated in A549-bearing mice. KEY FINDINGS CST and CAT can form self-assembled micelles, and have low value of critical micelle concentration. Notably, DOX-containing CST (D-CST) micelles demonstrated reduction-triggered drug release in glutathione-containing media. Further, reduction-responsive uptake of D-CST was observed in A549 cells. In addition, D-CST induced stronger cytotoxicity (P < 0.05) than DOX-loaded CAT (D-CAT) against A549 and AGS cells. Moreover, D-CST exhibited significantly stronger antitumour activity in A549-bearing nude mice than doxorubicin hydrochloride and D-CAT. CONCLUSIONS The reduction-responsive CST micelles enhanced the DOX effect at tumour site and controlled drug release.
Collapse
Affiliation(s)
- Tamara Davidovic
- Department of Internal Medicine III (Nephrology and Dialysis), Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Judith Schimpf
- Department of Internal Medicine III (Nephrology and Dialysis), Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Hannelore Sprenger-Mähr
- Department of Internal Medicine III (Nephrology and Dialysis), Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Armin Abbassi-Nik
- Department of Internal Medicine III (Nephrology and Dialysis), Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Afschin Soleiman
- Pathology, Cytodiagnostics and Molecular Pathology, Hall in Tirol, Austria
| | - Emanuel Zitt
- Department of Internal Medicine III (Nephrology and Dialysis), Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| | - Karl Lhotta
- Department of Internal Medicine III (Nephrology and Dialysis), Feldkirch Academic Teaching Hospital, Feldkirch, Austria
| |
Collapse
|
24
|
Chen H, Shi T, Wang Y, Liu Z, Liu F, Zhang H, Wang X, Miao Z, Liu B, Wan M, Mao C, Wei J. Deep Penetration of Nanolevel Drugs and Micrometer-Level T Cells Promoted by Nanomotors for Cancer Immunochemotherapy. J Am Chem Soc 2021; 143:12025-12037. [PMID: 34320319 DOI: 10.1021/jacs.1c03071] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The ability of nanomotors to promote the deep penetration of themselves and the loaded drugs in diseased tissues has been proposed and confirmed. However, whether such motion behavior of the nanomotors can also promote deep penetration of micrometer-sized immune cells in the diseased microenvironment, which is important for the immunotherapy of some diseases, has not been mentioned. Herein, we construct a nitric oxide (NO)-driven nanomotor that can move in the tumor microenvironment, focusing on its motion behavior and the role of NO, the beneficial product released during movement from this kind of nanomotor, in regulating the infiltration behavior and activity of immune cells. It can be found that the drug-loaded nanomotors with both NO-releasing ability and motility can promote the normalization of the tumor vasculature system and the degradation of the intrinsic extracellular matrix (ECM), which can significantly improve the tumor infiltration ability of T cells in vivo. The efficiency of T-cell infiltration in tumor tissue in vivo increased from 2.1 to 28.2%. Both subcutaneous and intraperitoneal implantation tumor models can validate the excellent antitumor effect of drug-loaded NO-driven nanomotors. This combination of motility of the power source from nanomotors and their physiological function offers a design idea for therapeutic agents for the future immunotherapy of many diseases.
Collapse
Affiliation(s)
- Huan Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Tao Shi
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing University, Nanjing 210008, China
| | - Yue Wang
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing University, Nanjing 210008, China
| | - Zhiyong Liu
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fangcen Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing University, Nanjing 210008, China
| | - Huanyu Zhang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xingwen Wang
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Zhuoyue Miao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing University, Nanjing 210008, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Jia Wei
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing University, Nanjing 210008, China.,Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210008, China
| |
Collapse
|
25
|
Li X, Zhang J, Liu P. Facile one-pot synthesis of amphiphilic acid/hypoxia co-triggered degradable diblock polyprodrug for tumor selective drug delivery. Int J Pharm 2021; 606:120941. [PMID: 34324987 DOI: 10.1016/j.ijpharm.2021.120941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 11/15/2022]
Abstract
More precise drug release is expected by conjugating the drug structural units in the polyprodrugs with dynamic covalent bonds responding to different stimuli. Here, amphiphilic acid/hypoxia co-triggered degradable diblock polyprodrug was designed via a facile one-pot method with drug content of 78.6% (1.22 mmol/g) and relatively molecular weight of 2.08 × 104, by condensation polymerization of acid-sensitive dimer of doxorubicin (D-DOXADH) with 2-iminothiolane, in presence of PEGylated D-DOXADH as end capping reagent for the PEGylation. Polyprodrug nanoparticles were easily obtained with mean hydrodynamic diameter of 177.6 ± 8.9 nm via self-assembly, which showed excellent acid/hypoxia co-triggered degradation and drug release performance. The ideal tumor selective cytotoxicity and enhanced antitumor efficacy were revealed with the in vitro cellular experiments. Such features make the proposed amphiphilic acid/hypoxia co-triggered degradable diblock polyprodrug a promising candidate for tumor chemotherapy.
Collapse
Affiliation(s)
- Xinming Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Juan Zhang
- Lanzhou University Hospital, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
26
|
Chen X, Jin R, Jiang Q, Bi Q, He T, Song X, Barz M, Ai H, Shuai X, Nie Y. Delivery of siHIF-1α to Reconstruct Tumor Normoxic Microenvironment for Effective Chemotherapeutic and Photodynamic Anticancer Treatments. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100609. [PMID: 34032365 DOI: 10.1002/smll.202100609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/17/2021] [Indexed: 06/12/2023]
Abstract
The tumor hypoxic microenvironment not only induces genetic and epigenetic changes in tumor cells, immature vessels formation for oxygen demand, but also compromises the efficiency of therapeutic interventions. On the other hand, conventional therapeutic approaches which kill tumor cells or destroy tumor blood vessels to block nutrition and oxygen supply usually facilitate even harsher microenvironment. Thus, simultaneously relieving the strained response of tumor cells and blood vessels represents a promising strategy to reverse the adverse tumor hypoxic microenvironment. In the present study, an integrated amphiphilic system (RSCD) is designed based on Angiotensin II receptor blocker candesartan for siRNA delivery against the hypoxia-inducible factor-1 alpha (HIF-1α), aiming at both vascular and cellular "relaxation" to reconstruct a tumor normoxic microenvironment. Both in vitro and in vivo studies have confirmed that the hypoxia-inducible HIF-1α expression is down-regulated by 70% and vascular growth is inhibited by 60%. The "relaxation" therapy enables neovascularization with more complete and organized structures to obviously increase the oxygen level inside tumor, which results in a 50% growth inhibition. Moreover, reconstruction of tumor microenvironment enhances tumor-targeted drug delivery, and significantly improves the chemotherapeutic and photodynamic anticancer treatments.
Collapse
Affiliation(s)
- Xiaobing Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qian Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qunjie Bi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Ting He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xu Song
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Matthias Barz
- Leiden Academic Center for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
- Institute of Organic Chemistry, Johannes Gutenberg-University, Mainz Duesbergweg 10-14, 55099, Mainz, Germany
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
27
|
Manzari MT, Shamay Y, Kiguchi H, Rosen N, Scaltriti M, Heller DA. Targeted drug delivery strategies for precision medicines. NATURE REVIEWS. MATERIALS 2021; 6:351-370. [PMID: 34950512 PMCID: PMC8691416 DOI: 10.1038/s41578-020-00269-6] [Citation(s) in RCA: 483] [Impact Index Per Article: 120.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/24/2020] [Indexed: 05/05/2023]
Abstract
Progress in the field of precision medicine has changed the landscape of cancer therapy. Precision medicine is propelled by technologies that enable molecular profiling, genomic analysis, and optimized drug design to tailor treatments for individual patients. Although precision medicines have resulted in some clinical successes, the use of many potential therapeutics has been hindered by pharmacological issues, including toxicities and drug resistance. Drug delivery materials and approaches have now advanced to a point where they can enable the modulation of a drug's pharmacological parameters without compromising the desired effect on molecular targets. Specifically, they can modulate a drug's pharmacokinetics, stability, absorption, and exposure to tumours and healthy tissues, and facilitate the administration of synergistic drug combinations. This Review highlights recent progress in precision therapeutics and drug delivery, and identifies opportunities for strategies to improve the therapeutic index of cancer drugs, and consequently, clinical outcomes.
Collapse
Affiliation(s)
- Mandana T. Manzari
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- These authors have contributed equally to this work
| | - Yosi Shamay
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- These authors have contributed equally to this work
| | - Hiroto Kiguchi
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- These authors have contributed equally to this work
| | - Neal Rosen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Maurizio Scaltriti
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel A. Heller
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
28
|
Zou L, Liu X, Li J, Li W, Zhang L, Fu C, Zhang J, Gu Z. Redox-sensitive carrier-free nanoparticles self-assembled by disulfide-linked paclitaxel-tetramethylpyrazine conjugate for combination cancer chemotherapy. Theranostics 2021; 11:4171-4186. [PMID: 33754055 PMCID: PMC7977472 DOI: 10.7150/thno.42260] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/17/2021] [Indexed: 12/21/2022] Open
Abstract
Rationale: Combinations of two or more therapeutic agents targeting different signaling pathways involved in tumor progression can have synergistic anticancer effects. However, combination chemotherapies are greatly limited by the different pharmacokinetics, tumor targeting, and cellular uptake capacities of the combined drugs. We have previously demonstrated the potential synergistic efficacy of paclitaxel (PTX) and the natural anti-angiogenic agent tetramethylpyrazine (TMP) for suppressing ovarian carcinoma growth. An efficient, facile, and smart nanosystem to deliver PTX and TMP simultaneously in vivo is greatly desired. Methods: We constructed a redox-sensitive nanosystem based on the amphiphilic PTX-ss-TMP conjugate, in which PTX and TMP are linked by a disulfide bond. We characterized the structure of the drug conjugate by 1H NMR and LC-MS, and then prepared PTX-ss-TMP NPs by a one-step nanoprecipitation method. We investigated the redox sensitivity, tumor-targeting ability, anticancer efficacy, and anti-angiogenesis activity of PTX-ss-TMP NPs in vitro and in vivo. Results: The amphiphilic PTX-ss-TMP conjugate readily self-assembled into stable nanoparticles in aqueous solution with a low critical association concentration of 1.35 µg/mL, well-defined spherical structure, small particle size (152 nm), high drug loading, redox-responsive drug release, high biocompatibility, and high storage stability. In cancer cells pretreated with GSH-OEt, PTX-ss-TMP NPs exhibited higher cytotoxicity, apoptosis rate, and cell-cycle arrest than monotherapy or combination therapy with free drugs, which was attributed to their improved cellular uptake and rapid intracellular drug release. Additionally, PTX-ss-TMP NPs also had a stronger anti-angiogenesis effect in HUVECs than free drug, which was mediated by VEGFR2-involved downstream signals. Finally, PTX-ss-TMP NPs showed tumor-specific accumulation and excellent antitumor activity in A2780 xenograft mice compared with free drug. Conclusions: These in vitro and in vivo results provide clear evidence that this redox-responsive carrier-free nanosystem with intrinsic amphiphilicity has great potential for combination cancer chemotherapy.
Collapse
|
29
|
Chintamaneni PK, Krishnamurthy PT, Pindiprolu SKSS. Polysorbate-80 surface modified nano-stearylamine BQCA conjugate for the management of Alzheimer's disease. RSC Adv 2021; 11:5325-5334. [PMID: 35423107 PMCID: PMC8694636 DOI: 10.1039/d1ra00049g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 01/13/2021] [Indexed: 01/31/2023] Open
Abstract
Acetylcholinesterase (AChE) inhibitors such as donepezil, galantamine and rivastigmine are used for the management of dementia in Alzheimer's Disease (AD). These drugs elevate endogenous acetylcholine (ACh) levels at the M1 muscarinic receptor in the brain to achieve therapeutic benefits. However, their side effects, such as nausea, vomiting, dizziness, insomnia, loss of appetite, altered heart rate, etc., are related to non-specific peripheral activation of M2-M5 muscarinic subtypes. It is logical, therefore, to develop drugs that selectively activate brain M1 receptors. Unfortunately, the orthosteric site homology among the receptor subtypes does not permit this approach. An alternative approach is to use positive allosteric modulator (PAM) of M1 receptors like benzyl quinolone carboxylic acid (BQCA). PAMs although devoid of M1 agonist activity, however, when bound, enhance the binding affinity of orthosteric ligand, Ach. The current challenge with PAMS is their low brain half-life, permeability, and higher elimination rates. This study reports active targeting of brain M1 receptors using surface modified nano lipid-drug conjugates (LDC) of M1 PAM, BQCA, to treat AD. Polysorbate-80 (P-80) surface modified stearylamine (SA)-BQCA conjugated nanoparticles (BQCA-SA-P80-NPs) were prepared by conjugating BQCA to SA, followed by the formation of nanoparticles (NPs) using P-80 by solvent injection method. The BQCA-SA-P80-NPs are near-spherical with a particle size (PS) of 166.62 ± 1.24 nm and zeta potential (ZP) of 23.59 ± 0.37 mV. In the in vitro cytotoxicity (SH-SY5Y cells) and hemolysis assays, BQCA-SA-P80-NPs, show acceptable safety and compatibility. In mice, Alzheimer's model, BQCA-SA-P80-NPs significantly prevent STZ induced changes in memory, neuronal Aβ1-42, p-Tau, APP, NF-κB, and BACE levels and neuronal cell death, when compared to untreated disease control and naïve BQCA treated group. Further, BQCA-SA-P80-NPs significantly improve the therapeutic efficacy of AChE inhibitor, donepezil (DPZ), indicating its potentiating effects. In vivo biodistribution studies in mice show selective accumulation of BQCA-SA-P80-NPs in the brain, suggesting an improved brain bioavailability and reduced peripheral side effects of BQCA. The study results demonstrate that BQCA-SA-P80-NPs can improve brain bioavailability and therapeutic efficacy of BQCA in AD.
Collapse
Affiliation(s)
- Pavan Kumar Chintamaneni
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education & Research) Ooty, The Nilgiris Tamil Nadu-643001 India +91-7598223850
- Department of Pharmaceutics, Raghavendra Institute of Pharmaceutical Education and Research (RIPER) Anantapuramu 51572 Andhra Pradesh India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education & Research) Ooty, The Nilgiris Tamil Nadu-643001 India +91-7598223850
| | - Sai Kiran S S Pindiprolu
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education & Research) Ooty, The Nilgiris Tamil Nadu-643001 India +91-7598223850
- Department of Pharmacology, Aditya Pharmacy College Surampalem East Godavari 533 437 Andhra Pradesh India
| |
Collapse
|
30
|
Yu Y, Xiang K, Xu M, Li Y, Cui J, Zhang L, Tang X, Zhu X, Qian L, Zhang M, Yang Y, Yu Q, Shen Y, Gan Z. Prodrug Nanomedicine Inhibits Chemotherapy-Induced Proliferative Burst by Altering the Deleterious Intercellular Communication. ACS NANO 2021; 15:781-796. [PMID: 33410660 DOI: 10.1021/acsnano.0c07113] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chemotherapy is one of the most commonly used clinical antitumor strategies. However, the therapy-induced proliferative burst, which always accompanies drug resistance and metastasis, has become a major obstacle during treatment. Except for some endogenous cellular or genetic mechanisms and some microenvironmental selection pressures, the intercellular connections in the tumor microenvironment (TME) are also thought to be the driving force for the acquired drug resistance and proliferative burst. Even though some pathway inhibitors or cell exempting strategies could be applied to partially avoid these unwanted communications, the complexity of the TME and the limited knowledge about those unknown detrimental connections might greatly compromise the efforts. Therefore, a more broad-spectrum strategy is urgently needed to relieve the drug-induced burst proliferation during various treatments. In this article, based on the possible discrepancies in metabolic activity between cells with different growth rates, several ester-bond-based prodrugs were synthesized. After screening, 7-ethyl-10-hyodroxycamptothecin-based prodrug nanoparticles were found to efficiently overcome the paclitaxel resistance, to selectively act on the malignantly proliferated drug-resistant cells and, furthermore, to greatly diminish the proliferative effect of common cytotoxic agents by blocking the detrimental intercellular connections. With the discriminating ability against malignant proliferating cells, the as-prepared prodrug nanomedicine exhibited significant anticancer efficacy against both drug-sensitive and drug-resistant tumor models, either by itself or by combining with highly potent nonselective chemotherapeutics. This work provides a different perspective and a possible solution for the treatment of therapy-induced burst proliferation.
Collapse
Affiliation(s)
- Yanting Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Keqi Xiang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mingzhi Xu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yuqiang Li
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiajunzi Cui
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lanqiong Zhang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaohu Tang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xianqi Zhu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lili Qian
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Meng Zhang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yan Yang
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qingsong Yu
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Youqing Shen
- Center for Bionanoengineering and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zhihua Gan
- Beijing Laboratory of Biomedical Materials, The State Key Laboratory of Organic-Inorganic Composites, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
31
|
Kumari R, Sunil D. A mechanistic insight into benefits of aggregation induced emissive luminogens in cancer. J Drug Target 2021; 29:592-608. [PMID: 33399029 DOI: 10.1080/1061186x.2020.1868479] [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: 10/22/2022]
Abstract
Exploration of advanced chemotheranostics that benefit from a combined in vivo strategy of cancer diagnosis and chemotherapy simultaneously is highly valued and will expose novel possibilities in modifying treatment and reduce side effects. In recent years, nanodrug delivery systems that incorporate aggregation-induced emissive luminogens (AIEgens) have been developed to track and monitor anticancer drug release, trace translocation processes and predict chemotherapeutic responses. There are several classes of AIEgen based chemotheranostics such us stimuli-responsive nanoprodrugs, pH-sensitive mesoporous silica nanocarriers, supramolecular polymer systems, drug encapsulated carriers, carrier-free nanodrugs, self-indicating drug delivery nanomachines and AIEgen-prodrug co-assembly. The present review conveys mechanistic insight into the benefits of AIEgens in the theranostic application by illustrating the recent breakthroughs in chemotheranostic nanomedicines that incorporate these unique fluorophores as signal reporters. The perspectives that can be further explored are also highlighted with the hope to instil more research interest in the advancement of AIE active cancer chemotheranostics for imaging and treatment in vivo.HIGHLIGHTSAggregation induced emissive materials (AIEgens) exhibit unique advantages over conventional luminogens for synergistic diagnosis and chemotherapy of cancer in vivo.The combination of AIE and nanotechnology offers an excellent platform to fabricate advanced chemotheranostics for cancer therapy.
Collapse
Affiliation(s)
- Rashmi Kumari
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| | - Dhanya Sunil
- Department of Chemistry, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
32
|
Sauraj, Kumar A, Kumar B, Kulshreshtha A, Negi YS. Redox-sensitive nanoparticles based on xylan-lipoic acid conjugate for tumor targeted drug delivery of niclosamide in cancer therapy. Carbohydr Res 2020; 499:108222. [PMID: 33401229 DOI: 10.1016/j.carres.2020.108222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 12/01/2020] [Accepted: 12/14/2020] [Indexed: 12/17/2022]
Abstract
In this study, novel redox-sensitive nanoparticles based on xylan-lipoic acid (Xyl-LA) conjugate were developed for tumor targeted delivery of niclosamide (Nic) in cancer therapy. The niclosamide loaded xylan-lipoic acid conjugate nanoparticles (Xyl-LA/Nic NPs) showed redox responsive behaviour in presence of reductive glutathione (GSH), which indicate their suitability for intracellular drug release. The obtained Xyl-LA/Nic NPs exhibited uniform particle size (196 ± 1.64 nm), high loading capacity (~28.6 wt %) and excellent blood compatibility. The anticancer activity of the Niclosamide and the Xyl-LA/Nic NPs against the colon carcinoma cell lines (HCT-15, Colo-320) were evaluated by MTT assay and the overall results indicate that the Xyl-LA/Nic NPs significantly enhanced the therapeutic efficiency of niclosamide in cancer therapy.
Collapse
Affiliation(s)
- Sauraj
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Saharanpur Campus, Paper Mill Road, Saharanpur, 247001, Uttar Pradesh, India
| | - Anuj Kumar
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Bijender Kumar
- Creative Research Center for Nanocellulose Future Composites, Department of Mechanical Engineering, Inha University, Incheon, 22212, South Korea
| | - Anurag Kulshreshtha
- Department of Paper Technology, Indian Institute of Technology Roorkee, Saharanpur Campus, Paper Mill Road, Saharanpur, 247001, Uttar Pradesh, India
| | - Yuvraj Singh Negi
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Saharanpur Campus, Paper Mill Road, Saharanpur, 247001, Uttar Pradesh, India.
| |
Collapse
|
33
|
Reduction-triggered di-block copolymer prodrug for high-performance long-acting tumor-selective killing. Colloids Surf B Biointerfaces 2020; 196:111368. [DOI: 10.1016/j.colsurfb.2020.111368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 01/19/2023]
|
34
|
Fucoidan-Doxorubicin Nanoparticles Targeting P-Selectin for Effective Breast Cancer Therapy. Carbohydr Polym 2020; 249:116837. [DOI: 10.1016/j.carbpol.2020.116837] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/29/2022]
|
35
|
Reduced Thiol Compounds – Induced Biosensing, Bioimaging Analysis and Targeted Delivery. CHINESE J CHEM 2020. [DOI: 10.1002/cjoc.202000320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Liu Y, Yang G, Jin S, Xu L, Zhao CX. Development of High-Drug-Loading Nanoparticles. Chempluschem 2020; 85:2143-2157. [PMID: 32864902 DOI: 10.1002/cplu.202000496] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/07/2020] [Indexed: 12/20/2022]
Abstract
Formulating drugs into nanoparticles offers many attractive advantages over free drugs including improved bioavailability, minimized toxic side effects, enhanced drug delivery, feasibility of incorporating other functions such as controlled release, imaging agents for imaging, targeting delivery, and loading more than one drug for combination therapies. One of the key parameters is drug loading, which is defined as the mass ratio of drug to drug-loaded nanoparticles. Currently, most nanoparticle systems have relatively low drug loading (<10 wt%), and developing methods to increase drug loading remains a challenge. This Minireview presents an overview of recent research on developing nanoparticles with high drug loading (>10 wt%) from the perspective of synthesis strategies, including post-loading, co-loading, and pre-loading. Based on these three different strategies, various nanoparticle systems with different materials and drugs are summarized and discussed in terms of their synthesis methods, drug loadings, encapsulation efficiencies, release profiles, stabilities, and their applications in drug delivery. The advantages and disadvantages of these strategies are presented with an objective of providing useful design rules for future development of high-drug-loading nanoparticles.
Collapse
Affiliation(s)
- Yun Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Guangze Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Song Jin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Letao Xu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| | - Chun-Xia Zhao
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland, 4072, Australia
| |
Collapse
|
37
|
Mehmood S, Wang L, Yu H, Haq F, Amin BU, Uddin MA, Fahad S, Haroon M, Shen D, Ni Z. Preparation of poly(cyclotriphosphazene-co-piperazine) nanospheres and their drug release behavior. INT J POLYM MATER PO 2020. [DOI: 10.1080/00914037.2020.1809407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Sahid Mehmood
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Fazal Haq
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Bilal-ul Amin
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Md Alim Uddin
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Shah Fahad
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Muhammad Haroon
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Di Shen
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| | - Zhipeng Ni
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, P.R. China
| |
Collapse
|
38
|
Zhang Y, Fang F, Li L, Zhang J. Self-Assembled Organic Nanomaterials for Drug Delivery, Bioimaging, and Cancer Therapy. ACS Biomater Sci Eng 2020; 6:4816-4833. [PMID: 33455214 DOI: 10.1021/acsbiomaterials.0c00883] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Over the past few decades, tremendous progress has been made in the development of engineering nanomaterials, which opened new horizons in the field of diagnosis and treatment of various diseases. In particular, self-assembled organic nanomaterials with intriguing features including delicate structure tailoring, facile processability, low cost, and excellent biocompatibility have shown outstanding potential in biomedical applications because of the enhanced permeability and retention (EPR) effect and multifunctional properties. In this review, we briefly introduce distinctive merits of self-assembled organic nanomaterials for biomedical applications. The main focus will be placed on summarizing recent advances in self-assembled organic nanomedicine for drug delivery, bioimaging, and cancer phototherapy, followed by highlighting a critical perspective on further development of self-assembled organic nanomaterials for future clinical translation. We believe that the above themes will appeal to researchers from different fields, including material, chemical, and biological sciences, as well as pharmaceutics.
Collapse
Affiliation(s)
- Yinfeng Zhang
- International Medical Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China
| | - Fang Fang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100811, P. R. China
| | - Li Li
- International Medical Center, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P. R. China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing 100811, P. R. China
| |
Collapse
|
39
|
Ma Y, Mou Q, Yan D, Zhu X. Engineering small molecule nanodrugs to overcome barriers for cancer therapy. VIEW 2020. [DOI: 10.1002/viw.20200062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- Yuan Ma
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Quanbing Mou
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering State Key Laboratory of Metal Matrix Composites Shanghai Jiao Tong University Shanghai China
| |
Collapse
|
40
|
Li S, Shan X, Wang Y, Chen Q, Sun J, He Z, Sun B, Luo C. Dimeric prodrug-based nanomedicines for cancer therapy. J Control Release 2020; 326:510-522. [PMID: 32721523 DOI: 10.1016/j.jconrel.2020.07.036] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 01/10/2023]
Abstract
With the rapid development of conjugation chemistry and biomedical nanotechnology, prodrug-based nanosystems (PNS) have emerged as promising drug delivery nanoplatforms. Dimeric prodrug, as an emerging branch of prodrug, has been widely investigated by covalently conjugating two same or different drug molecules. In recent years, great progress has been made in dimeric prodrug-based nanosystems (DPNS) for cancer therapy. Many advantages offered by DPNS have significantly facilitated the delivery efficiency of anticancer drugs, such as high drug loading capacity, favorable pharmacokinetics, tumor stimuli-sensitive drug release and facile combination theranostics. Given the rapid developments in this field, we here outline the latest updates of DPNS in cancer treatment, focusing on dimeric prodrug-encapsulated nanosystems, dimeric prodrug-nanoassemblies and tumor stimuli-responsive DPNS. Moreover, the design principle, advantages and challenges of DPNS for clinical cancer therapy are also highlighted.
Collapse
Affiliation(s)
- Shumeng Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xinzhu Shan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuequan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Qin Chen
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, PR China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Cong Luo
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
41
|
Wang Z, Chen J, Little N, Lu J. Self-assembling prodrug nanotherapeutics for synergistic tumor targeted drug delivery. Acta Biomater 2020; 111:20-28. [PMID: 32454086 PMCID: PMC7245299 DOI: 10.1016/j.actbio.2020.05.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/27/2020] [Accepted: 05/18/2020] [Indexed: 01/08/2023]
Abstract
Self-assembling prodrugs represents a robust and effective nanotherapeutic approach for delivering poorly soluble anticancer drugs. With numerous intrinsic advantages, self-assembling prodrugs possess the maximum drug loading capacity, controlled drug release kinetics, prolonged blood circulation, and preferential tumor accumulation based on the enhanced permeability and retention (EPR) effect. These prodrug conjugates allow for efficient self-assembly into nanodrugs with the potential of encapsulating other therapeutic agents that have different molecular targets, enabling simultaneous temporal-spatial release of drugs for synergistic antitumor efficacy with reduced systemic side effects. The aim of this review is to summarize the recent progress of self-assembling prodrug cancer nanotherapeutics that are made through conjugating therapeutically active agents to Polyethylene glycol, Vitamin E, or drugs with different physicochemical properties via rational design, for synergistic tumor targeted drug delivery. Statement of Significance All current FDA-approved nanomedicines use inert biomaterials as drug delivery carriers. These biomaterials lack any therapeutic potential, contributing not only to the cost, but may also elicit severe unfavorable adverse effects. Despite the reduction in toxicity associated with the payload, these nanotherapeutics have been met with limited clinical success, likely due to the monotherapy regimen. The self-assembling prodrug (SAP) has been emerging as a powerful platform for enhancing efficacy through co-delivering other therapeutic modalities with distinct molecular targets. Herein, we opportunely present a comprehensive review article summarizing three unique approaches of making SAP for synergistic drug delivery: pegylation, vitamin E-derivatization, and drug-drug conjugation. These SAPs may inevitably pave the way for developing more efficacious, clinically translatable, combination cancer nanotherapies.
Collapse
|
42
|
Hybrid micelles based on Pt (IV) polymeric prodrug and TPGS for the enhanced cytotoxicity in drug-resistant lung cancer cells. Colloids Surf B Biointerfaces 2020; 195:111256. [PMID: 32682273 DOI: 10.1016/j.colsurfb.2020.111256] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/21/2020] [Accepted: 07/10/2020] [Indexed: 12/17/2022]
Abstract
Multidrug resistance (MDR) is a primary cause of failure in oncotherapy and interest is growing in the design of multi-stimuli responsive nano-carriers to synergistically deliver chemotherapeutic agents and P-gp inhibitors to reverse MDR. The hybrid micelles based on a Platinum (IV)-coordinate polymeric prodrugs and TPGS were developed to improve chemotherapy and reduce side effects. The pH/redox dual-sensitive polymers were synthesized by condensation polymerization using ortho ester monomer and diamminedichlorodisuccinatoplatinum (DSP). The hybrid micelles possessed uniform size (38 nm) and displayed good stability in various physiological conditions. In contrast, in vitro drug release profiles indicated that these micelles could be completely depolymerized under acidic and reducing environment, thereby more than 80 % cisplatin were released within 12 h at pH 5.0 plus 10 mM DTT. More importantly, a large amount of TPGS released simultaneously could effectively inhibit the function of drug efflux pumps, which significantly enhanced the cytotoxicity of cisplatin against A549/DDP cells. The growth inhibition rate of micelles on A549/DDP multicellular spheroids was 79.5 %, while that of free cisplatin was only 6.8 %. Therefore, these hybrid micelles are promising in overcoming tumor MDR and worth doing further research in vivo and extend to other therapeutic agents.
Collapse
|
43
|
Charge-reversible lipid derivative: A novel type of pH-responsive lipid for nanoparticle-mediated siRNA delivery. Int J Pharm 2020; 585:119479. [DOI: 10.1016/j.ijpharm.2020.119479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 12/15/2022]
|
44
|
pH-Responsive supramolecular DOX-dimer based on cucurbit[8]uril for selective drug release. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2019.10.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
45
|
Li J, Li X, Pei M, Liu P. Acid-labile anhydride-linked doxorubicin-doxorubicin dimer nanoparticles as drug self-delivery system with minimized premature drug leakage and enhanced anti-tumor efficacy. Colloids Surf B Biointerfaces 2020; 192:111064. [PMID: 32387860 DOI: 10.1016/j.colsurfb.2020.111064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 01/21/2023]
Abstract
Acid-labile anhydride-linked doxorubicin-doxorubicin dimers (D-DOX) were designed as doxorubicin-doxorubicin conjugate-based drug self-delivery systems (DSDSs) with high drug content for tumor intracellular pH-triggered release, by conjugating doxorubicin (DOX) with various anhydrides, such as maleic anhydride (MAH), succinic anhydride (suc), and 2,3-dimethylmaleic anhydride (DMMAH). With the similar diameter of about 200 nm, the D-DOXMAH showed better pH-triggered DOX release and was thus selected for the further investigation. The D-DOX-5 nanoparticles with desirable average hydrodynamic diameter (Dh) of 162 nm and high drug content of 51.20% were obtained via self-assembly by a facile dialysis technique, with the PEGylated dimer (D-DOXMAH-S-PEG) as surfactant. The cumulative DOX release from the proposed D-DOX nanoparticles reached 40.6% within 36 h in the simulated tumor intracellular acidic micro-environment, while the premature drug leakage was only 4.5% in the simulated normal physiological medium. The MTT results indicated the proposed DSDS possessed an enhanced anti-tumor efficacy for the HepG2 cancer cell than the free DOX.
Collapse
Affiliation(s)
- Jiagen Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Mingliang Pei
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
46
|
Zong L, Wang Y, Qiao P, Yu K, Hou X, Wang P, Zhang Z, Pang X, Pu X, Yuan Q. Reduction-sensitive poly(ethylene glycol)-polypeptide conjugate micelles for highly efficient intracellular delivery and enhanced antitumor efficacy of hydroxycamptothecin. NANOTECHNOLOGY 2020; 31:165102. [PMID: 31899896 DOI: 10.1088/1361-6528/ab6749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The non-specific biodistribution of traditional chemotherapeutic drugs against tumors is the key factor that causes systemic toxicity and hinders their clinical application. In this study, a reduction-sensitive polymer conjugate micelle was manufactured to achieve tumor-specific targeting, reduce toxic side-effects and improve anti-tumor activity of a natural anti-cancer drug, hydroxycamptothecin (HCPT). Therefore, HCPT was conjugated with methoxy-poly(ethylene glycol)-poly(β-benzyl-L-aspartate) (mPEG-PBLA) by a disulfide bond or succinate bond for the first time to obtain the mPEG-PBLA-SS-HCPT (PPSH) and mPEG-PBLA-CC-HCPT (PPCH) that would form micelles after high-speed agitation and dialysis. The PPSH micelles showed an average particle size of 126.3 nm, a low polydispersity index of 0.209, and a negative surface charge of -21.1 mV zeta potential. Transmission electron microscopy showed the PPSH micelles to have spherical morphology. PPSH had a low critical micelle concentration of 1.29 μg ml-1 with high dilution stability, storage stability and reproducibility. Moreover, the particle size of the PPSH micelles had no significant change after incubation with rat plasma for 72 h, probably resulting in high long circulation in the blood. The PPSH micelles showed significant reduction sensitivity to glutathione. Their sizes increased by 403.2 nm after 24 h post-incubation, and 87.6% drug release was achieved 48 h post-incubation with 40 mM glutathione solutions. The PPSH micelles showed stronger inhibition of HepG2 cells in vitro and growth of H-22 tumor in vivo than the PPCH and HCPT solutions after intravenous injection. The accumulation of PPSH micelles in the tumor tissue contributed to the high anti-tumor effect with little side-effect on the normal tissues. The reduction-sensitive PPSH micelles were a promising carrier of HCPT and other poorly soluble anti-cancer drugs.
Collapse
|
47
|
Li N, Guo W, Li Y, Zuo H, Zhang H, Wang Z, Zhao Y, Yang F, Ren G, Zhang S. Construction and anti-tumor activities of disulfide-linked docetaxel-dihydroartemisinin nanoconjugates. Colloids Surf B Biointerfaces 2020; 191:111018. [PMID: 32304917 DOI: 10.1016/j.colsurfb.2020.111018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 12/19/2022]
Abstract
Co-delivery of anti-tumor agents with outstanding stimulus-triggered drug release in tumor cells, especially with the aid of nanotechnology, provided the possibility to enhance delivery efficiency for targeting tumor cells and antitumor efficacy. In this paper, docetaxel-dihydroartemisinin nanoconjugates linked by disulfide bond were designed to increase co-delivery and anti-tumor efficacy. Docetaxel and dihydroartemisinin were synthesized using two-step reaction and furtherly assembled to nanoconjugates. Nanoprescription was optimized to evaluate its physicochemical properties. In vitro anti-tumor activities of nanoformulation were assessed by MTT. The flow cytometry was adopted to analyze cell apoptosis and cell cycle arrest. The wound healing assay was used to evaluate antimigratory-property. In vivo pharmacokinetic and pharmacodynamic studies were investigated in rats and 4T1 bearing Balb/c mice model after intravenous injection, respectively. The chemical structure of conjugate was confirmed. The prepared nanoparticles possessed uniform size distribution (172.10 ± 1.70 nm, PDI 0.05 ± 0.01), was stable during storage period, sustained release profiles and sensitive reduction responsiveness. MTT assay indicated that the toxicity of nanoconjugates was slightly weak. Flow cytometry studies showed that nanoconjugates could promote early apoptosis significantly and mainly arose from G0/G1 phase. The wound healing assay provided an obvious antimetastatic potential of nanoparticles in 4T1 cells. The result of pharmacokinetic study suggested that nanoconjugates exhibited higher exposure levels. In vivo pharmacodynamic research showed that mice treated with docetaxel-dihydroartemisinin nanoconjugates had lower systemic toxicity and higher survival ratio than those of control groups. This potential of nanoconjugates was developed as a novel nanoplateform to treat tumor.
Collapse
Affiliation(s)
- Ning Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wenju Guo
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yujie Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hengtong Zuo
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huihui Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhaoyun Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongdan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fan Yang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guolian Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
48
|
Redox responsive xylan-SS-curcumin prodrug nanoparticles for dual drug delivery in cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 107:110356. [DOI: 10.1016/j.msec.2019.110356] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 10/17/2019] [Accepted: 10/21/2019] [Indexed: 02/05/2023]
|
49
|
Li J, Li X, Liu P. Synthesis of Acid-Labile Poly(Doxazolidine) as a Polyprodrug with an Ultra-High Drug Content for Self-Delivery of High-Performance Chemotherapeutics. Mol Pharm 2020; 17:710-716. [PMID: 31910025 DOI: 10.1021/acs.molpharmaceut.9b00972] [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: 11/28/2022]
Abstract
Drug self-delivery systems (DSDSs) have attracted intense attention due to their high drug content. However, their practical application still suffers from their premature drug leakage, slow drug release, and/or low antitumor efficacy of the released small molecular drugs. Here, acid-labile poly(Doxazolidine) (P(Doxaz)) is designed as a polyprodrug for the self-delivery of high antitumor chemotherapeutics (Doxazolidine (Doxaz)), with an ultrahigh Doxaz content of 92.45%. The P(Doxaz) nanoparticles could completely degrade into Doxaz within 10 h in the simulated tumor intracellular microenvironment, with a low drug leakage of 12.9% over 12 h in the normal physiological media. Owing to the ultrahigh drug content, fast acid-triggered degradation and drug release, and high antitumor efficacy of Doxaz, the proposed DSDS possesses an enhanced antiproliferation efficacy compared to the free DOX, demonstrating its potential in future tumor treatments.
Collapse
Affiliation(s)
- Jiagen Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering , Lanzhou University , Lanzhou 730000 , China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering , Lanzhou University , Lanzhou 730000 , China
| | - Peng Liu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering , Lanzhou University , Lanzhou 730000 , China
| |
Collapse
|
50
|
Hu H, Zhou H, Zhen Z, Wu Z, Zhang R, Xu D. Methoxylpoly(ethylene glycol)‐retinoic acid Micelles Loaded with Dimethylcurcumin for Efficient Castration‐Resistant Prostate Cancer Therapy. ChemistrySelect 2019. [DOI: 10.1002/slct.201902562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Hang Hu
- School of Pharmaceutical Engineering and Life ScienceChangzhou University Changzhou 213164 P. R. China
| | - Huan Zhou
- Center for Health Science and EngineeringTianjin Key Laboratory of Materials Laminating Fabrication and Interface Control TechnologySchool of Materials Science and EngineeringHebei University of Technology Tianjin 300130 P. R. China
- School of Mechanical EngineeringJiangsu University of Technology, Changzhou Jiangsu 213001 P. R. China
| | - Zihan Zhen
- School of Pharmaceutical Engineering and Life ScienceChangzhou University Changzhou 213164 P. R. China
| | - Zhe Wu
- School of Pharmaceutical Engineering and Life ScienceChangzhou University Changzhou 213164 P. R. China
| | - Rong Zhang
- School of Materials Science & EngineeringChangzhou University Changzhou 213164 P. R. China
| | - Defeng Xu
- School of Pharmaceutical Engineering and Life ScienceChangzhou University Changzhou 213164 P. R. China
- National & Local Joint Engineering Research Center for High-efficiency Refining and High-quality Utilization of BiomassChangzhou University Changzhou 213164 P. R. China
| |
Collapse
|