1
|
Ye F, Li Q, Huang L, Liao N. Reliable high-PAP-1-loaded polymeric micelles for cancer therapy: preparation, characterization, and evaluation of anti-tumor efficacy. Drug Deliv 2025; 32:2490269. [PMID: 40207975 PMCID: PMC11986873 DOI: 10.1080/10717544.2025.2490269] [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: 12/13/2024] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/11/2025] Open
Abstract
The mitochondrial potassium channel Kv1.3 is a critical therapeutic target, as its blockade induces cancer cell apoptosis, highlighting its therapeutic potential. PAP-1, a potent and selective membrane-permeant Kv1.3 inhibitor, faces solubility challenges affecting its bioavailability and antitumor efficacy. To circumvent these challenges, we developed a tumor-targeting drug delivery system by encapsulating PAP-1 within pH-responsive mPEG-PAE polymeric micelles. These self-assembled micelles exhibited high entrapment efficiency (91.35%) and drug loading level (8.30%). As pH decreased, the micelles exhibited a significant increase in particle size and zeta potential, accompanied by a surge in PAP-1 release. Molecular simulations revealed that PAE's tertiary amine protonation affected the self-assembly process, modifying hydrophobicity and resulting in larger, loosely packed particles. Furthermore, compared to free PAP-1 or PAP-1 combined with MDR inhibitors, PAP-1-loaded micelles significantly enhanced cytotoxicity and apoptosis induction in Jurkat and B16F10 cells, through mechanisms involving decreased mitochondrial membrane potential and elevated caspase-3 activity. In vivo, while free PAP-1 failed to reduce tumor size in a B16F10 melanoma mouse model, PAP-1-loaded micelles substantially suppressed tumors, reducing volume by up to 94.26%. Fluorescent-marked micelles effectively accumulated in mouse tumors, confirming their targeting efficiency. This strategy holds promise for significantly improving PAP-1's antitumor efficacy in tumor therapy.
Collapse
Affiliation(s)
- Fang Ye
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, P. R. China
| | - Qi Li
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, P. R. China
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, P. R. China
| | - Longping Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, P. R. China
- Clinical Laboratory, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, P.R. China
| | - Naikai Liao
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, P. R. China
| |
Collapse
|
2
|
Summer M, Hussain T, Ali S, Khan RRM, Muhammad G, Liaqat I. Exploring the underlying modes of organic nanoparticles in diagnosis, prevention, and treatment of cancer: a review from drug delivery to toxicity. INT J POLYM MATER PO 2025; 74:829-845. [DOI: 10.1080/00914037.2024.2375337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore
| | - Tauqeer Hussain
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore
| | - Rana Rashad Mahmood Khan
- Department of Chemistry, Government College University Lahore, Faculty of Chemistry and Life Sciences
| | - Gulzar Muhammad
- Department of Chemistry, Government College University Lahore, Faculty of Chemistry and Life Sciences
| | - Iram Liaqat
- Microbiology Lab, Department of Zoology, Government College University Lahore
| |
Collapse
|
3
|
Chatterjee S, Tyagi M, Ghosh A, Chandwadkar P, Nagar S, Mor J, Wadawale AP, Acharya C, Patro BS, Goswami D. Robust Hydrogen-Bonded Organic Framework for pH-Responsive Drug Delivery. Biomacromolecules 2025. [PMID: 40490986 DOI: 10.1021/acs.biomac.5c00316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
Hydrogen-bonded organic frameworks (HOFs), by virtue of their low toxicity, wide substrate tolerance, porosity, and regenerative and biocompatible traits, are an emerging class of porous polymeric materials that show great potential for intracellular delivery of chemotherapeutics. However, stability issues coupled with controlled release of a drug under desired stimuli have restricted their therapeutic potential. In the present study, based on density functional theory calculations predicting strong noncovalent interactions between the H-bonded 4,4',4″-(1,3,5-triazine-2,4,6-triyl)tribenzoic acid (H3TATB) dimer and the chemotherapeutic drug doxorubicin (Dox), we prepared a robust and reticular nanoplatform Dox@nano-HOF 1, which released Dox at low pH, making it an ideal carrier for targeted drug delivery to tumor sites sparing the normal tissues. Additionally, the cytotoxic and apoptosis assays confirmed the efficacy of the nanocarrier in inducing dose-dependent cell death in cancer cells. This study opens up new and hitherto unexplored avenues for the use of robust HOFs in pH-responsive delivery of anticancer therapeutics.
Collapse
Affiliation(s)
- Sucheta Chatterjee
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Mrityunjay Tyagi
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Ayan Ghosh
- Laser and Plasma Technology Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Pallavi Chandwadkar
- Molecular Biology Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Suryakant Nagar
- Product Development Division, Bhabha Atomic Research Centre, Trombay, Trombay, Mumbai 400085, India
| | - Jaideep Mor
- Radiochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Amey P Wadawale
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Celin Acharya
- Molecular Biology Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Birija S Patro
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Dibakar Goswami
- Bio-Organic Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
4
|
Liu JJJ, Liu D, To SKY, Wong AST. Exosomes in cancer nanomedicine: biotechnological advancements and innovations. Mol Cancer 2025; 24:166. [PMID: 40481526 PMCID: PMC12144782 DOI: 10.1186/s12943-025-02372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Accepted: 05/28/2025] [Indexed: 06/11/2025] Open
Abstract
Exosomes, as natural intercellular messengers, are gaining prominence as delivery vehicles in nanomedicine, offering a superior alternative to conventional synthetic nanoparticles for cancer therapeutics. Unlike lipid, polymer, or metallic nanoparticles, which often face challenges related to immunogenicity, targeting precision, and off-tumor toxicity, exosomes can effectively encapsulate a diverse range of therapeutic agents while exhibiting low toxicity, favorable pharmacokinetics, and organotropic properties. This review examines recent advancements in exosome bioengineering over the past decade. Innovations such as microfluidics-based platforms, nanoporation, fusogenic hybrids, and genetic engineering have significantly improved loading efficiencies, production scalability, and pharmacokinetics of exosomes. These advancements facilitate tumor-specific cargo delivery, resulting in substantial improvements in retention and efficacy essential for clinical success. Moreover, enhanced biodistribution, targeting, and bioavailability-through strategies such as cell selection, surface modifications, membrane composition alterations, and biomaterial integration-suggests a promising future for exosomes as an ideal nanomedicine delivery platform. We also highlight the translational impact of these strategies through emerging clinical trials. Additionally, we outline a framework for clinical translation that focuses on: cargo selection, organotropic cell sourcing, precision loading methodologies, and route-specific delivery optimization. In summary, this review emphasizes the potential of exosomes to overcome the pharmacokinetic and safety challenges that have long impeded oncology drug development, thus enabling safer and more effective cancer treatments.
Collapse
Affiliation(s)
- Jacky J J Liu
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| | - Duanrui Liu
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Sally K Y To
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, 17 W, Hong Kong Science and Technology Parks, New Territories, Hong Kong.
| | - Alice S T Wong
- School of Biological Sciences, University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong.
| |
Collapse
|
5
|
Lozano-García M, Dikici E, Bilbao D, Mohan P, Deo S, Daunert S. Multifunctional delivery strategies and nanoplatforms of SN-38 in cancer therapeutics. J Control Release 2025; 384:113937. [PMID: 40490199 DOI: 10.1016/j.jconrel.2025.113937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 06/03/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
SN-38 or 7-ethyl-10-hydroxycamptothecin is the active metabolite of irinotecan, a widely used chemotherapeutic agent for the treatment of colorectal, pancreatic, lung, breast, gastric, esophageal, hepatocellular, ovarian, brain, leukemia, and lymphoma malignancies. SN-38's antitumoral effect is 100 to 1000 times more potent than that of irinotecan. However, its clinical application is hindered by its poor solubility and chemical instability. To circumvent these challenges and avoid systemic toxicities, such as myelosuppression and diarrhea, several SN-38 delivery systems have been explored. In that regard, formulations based on targeted, controlled and tumor-responsive release of SN-38 have demonstrated to enhance its antitumoral effects and reduce the associated systemic toxicities by limiting the pharmacological activity to the desired tumor location. To this end, prodrugs, conjugates, nanoparticles, dendrimers, or lipid-based strategies for SN-38 delivery have been used. Most recently, multifunctional approaches have emerged as an attractive alternative to develop SN-38 delivery systems, combining several strategies in a single formulation, i.e., encapsulating nanocarriers, tumor-targeting ligands, stimuli-responsive elements, optimal linkers, drug combinations or bioimaging agents. Despite their therapeutic advantages, multifunctional delivery systems often face challenges concerning their clinical translation compared to conventional therapies, such as biocompatibility, scalability and cost-effectiveness issues. The aim of this work is to review the most recent progress that has been made in the development and assessment of multifunctional delivery systems for cancer treatment.
Collapse
Affiliation(s)
- Mercedes Lozano-García
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA
| | - Daniel Bilbao
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Prasoon Mohan
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; Vascular and Interventional Radiology, University of Miami Health System, Miami, FL 33136, USA
| | - Sapna Deo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
6
|
Ai Z, Liu B, Chen J, Zeng X, Wang K, Tao C, Chen J, Yang L, Ding Q, Zhou M. Advances in nano drug delivery systems for enhanced efficacy of emodin in cancer therapy. Int J Pharm X 2025; 9:100314. [PMID: 39834843 PMCID: PMC11743866 DOI: 10.1016/j.ijpx.2024.100314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/05/2025] Open
Abstract
Cancer remains one of the leading causes of death worldwide, highlighting the urgent need for novel antitumor drugs. Natural products have long been a crucial source of anticancer agents. Among these, emodin (EMO), a multifunctional anthraquinone compound, exhibits significant anticancer effects but is hindered in clinical applications by challenges such as low solubility, rapid metabolism, poor bioavailability, and off-target toxicity. Nano drug delivery systems offer effective strategies to overcome these limitations by enhancing the solubility, stability, bioavailability, and targeting ability of EMO. While substantial progress has been made in developing EMO-loaded nanoformulations, a comprehensive review on this topic is still lacking. This paper aims to fill this gap by providing an overview of recent advancements in nanocarriers for EMO delivery and their anticancer applications. These carriers include liposomes, nanoparticles, polymeric micelles, nanogels, and others, with nanoparticle-based formulations being the most extensively explored. Nanoformulations encapsulating EMO have demonstrated promising therapeutic results against various cancers, particularly breast cancer, followed by liver and lung cancers. We systematically summarize the preparation methods, materials, and physicochemical properties of EMO-loaded nanopreparations, underscoring key findings on how nanotechnology improves the anticancer efficacy of EMO. This review provides valuable insights for researchers engaged in developing nano delivery systems for anticancer drugs.
Collapse
Affiliation(s)
- Zhenghao Ai
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Bingyao Liu
- Department of Radiology, West China Hospital Sichuan University Jintang Hospital, Chengdu, China
| | - Junyan Chen
- Department of Cardiothoracic Surgery, Luzhou People's Hospital, Luzhou, China
| | - Xinhao Zeng
- Department of Pediatric Surgery, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, China
| | - Ke Wang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chao Tao
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jing Chen
- Department of Clinical Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Liuxuan Yang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Qian Ding
- Department of Clinical Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
7
|
Pourbakhsh M, Jabraili M, Akbari M, Jaymand M, Jahanban Esfahlan R. Poloxamer-based drug delivery systems: Frontiers for treatment of solid tumors. Mater Today Bio 2025; 32:101727. [PMID: 40275957 PMCID: PMC12018049 DOI: 10.1016/j.mtbio.2025.101727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Pluronics or poloxamers are a type of triblock copolymer. These non-ionic molecules consist of a hydrophobic block embedded in two hydrophilic parts. Pluronics have become favorite materials for use in the field of biomedical research due to having favorable physicochemical and biological properties such as amphiphilicity, solubility in ionic and non-ionic solutions, biocompatibility, biodegradability, self-assembly and low toxicity. The scope of these applications can vary from tissue engineering to drug delivery. One of the important uses of pluronics is to deliver drugs to various cancer cells. Herein we first provide an overview on variety of ploronic biomaterials. And then intensively evaluate their potential as drug delivery systems (DDSs) for treatment of solid tumors with special focus on breast cancers. After explaining the pros and cons of pluronics, the current status in clinical settings and future prospects are highlighted.
Collapse
Affiliation(s)
- Mehdi Pourbakhsh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Jabraili
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Jaymand
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Rana Jahanban Esfahlan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Zhang H, Du Y, Liu X, Gao R, Xu X, Hou X, Liu Z, Xin P, Wu W, Liu T, Zhao Y, Zhou H, Jiang Y. Enhancing curcumol delivery through PD-1 targeted nanocarriers: A novel therapeutic approach for prostate cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156595. [PMID: 40220412 DOI: 10.1016/j.phymed.2025.156595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Prostate cancer is a prevalent form of cancer that impacts men on a global scale, and its treatment faces challenges such as tumor metastasis, immune resistance, and epigenetic abnormalities. Most current research focuses on nanocarriers with a single function, but the dual mechanism of action-enhancing immune response and regulating EZH2 epigenetic modification-has not been reported. PURPOSE This study is the first to construct an engineered outer membrane vesicle (OMV) delivery system loaded with PD-1 antibody and Curcumol, combining two cutting-edge approaches: tumor immunotherapy and epigenetic regulation. We developed a nanocarrier system based on engineered OMVs (OMV-PD-1) to deliver the natural anticancer compound Curcumol, aiming to regulate epigenetic modifications and enhance tumor immune responses, thereby effectively inhibiting the proliferation and metastasis of prostate cancer cells. METHODS OMV-PD-1 was prepared using recombinant technology, and its characteristics were identified through the application of liquid chromatography-mass spectrometry (LC-MS), dynamic light scattering (DLS), and transmission electron microscopy (TEM). We assessed its antitumor activity against prostate cancer cells (PC3) in vitro and in vivo and explored its molecular mechanisms through RNA sequencing and gene set enrichment analysis (GSEA). RESULTS An outstanding encapsulation efficiency and a delayed drug release profile were evident in OMV-PD-1/Curcumol. In vitro experiments demonstrated that the system significantly inhibited PC3 cell migration (77.25 % inhibition) and invasion (73.03 % inhibition), and regulated histone methylation modifications (such as H3K9 and H3K27) by downregulating EZH2 gene expression. In vivo experiments confirmed its excellent tumor targeting in a humanized mouse model, significantly inhibiting tumor growth and enhancing immune responses, such as increased NK cell infiltration and elevated pro-inflammatory cytokine levels. CONCLUSION The OMV-PD-1/Curcumol delivery system developed in this study not only hinders the aggressive actions of prostate cancer cells by regulating epigenetic modifications but also significantly stimulates antitumor immune responses, offering a unique and readily implementable therapeutic avenue.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Yang Du
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Xujia Liu
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, 110134, China
| | - Ruxu Gao
- Department of Urology, The Cancer Hospital of Dalian University of Technology&Liaoning Cancer Hospital and institute, Shenyang, Liaoning Province 110042, China
| | - Xiaoxiao Xu
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Xiaolong Hou
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Zhenghua Liu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Weiwei Wu
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Tao Liu
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China
| | - Yue Zhao
- Department of Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, School of Life Sciences, China Medical University, Shenyang City, Liaoning Province 110122, China.
| | - Hongxu Zhou
- Department of Anesthesiology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China.
| | - Yuanjun Jiang
- Department of Urology, The First Hospital of China Medical University, NO.155 North Nanjing Street, Heping District, Shenyang, Liaoning Province 110001, China.
| |
Collapse
|
9
|
Sipos B, Rajab F, Katona G, Csóka I. Current insights into polymeric micelles for nasal drug delivery. Expert Opin Drug Deliv 2025:1-18. [PMID: 40420578 DOI: 10.1080/17425247.2025.2511962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 05/09/2025] [Accepted: 05/23/2025] [Indexed: 05/28/2025]
Abstract
INTRODUCTION The nasal administration route has gained peak interest in recent literature and as a noninvasive alternative for efficient drug delivery and increasing bioavailability of active substances. Technological challenges arise from the drug's physicochemical properties and the nasal mucosal barrier for which innovative particle engineering techniques must be implemented, such as using polymeric nanocarriers. AREAS COVERED This review deals with the importance of the nasal administration route and its connection to polymeric micelles as innovative nanocarriers. The period between 2015-2025 up to date was chosen to search for original research articles where polymeric micelles were applied nasally. The first part demonstrates the utilization of polymeric micelles, followed by a summary of how drug release and permeability can be achieved in the nasal cavity and through the nasal epithelium. The second part reviews the studies conducted on this matter. EXPERT OPINION The nasal route could be superior to perform as a suitable alternative to conventional routes. Multiple studies have already demonstrated that the main advantages lie in the nose-to-brain drug delivery pathway, which can be conquered via adequately formulated polymeric micelles. As an innovative solution, vaccine delivery is also of great potential by combining the advantages of the delivery route and the polymeric nanocarriers.
Collapse
Affiliation(s)
- Bence Sipos
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Fatima Rajab
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Gábor Katona
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| | - Ildikó Csóka
- Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, Szeged, Hungary
| |
Collapse
|
10
|
Mohamed SMM, Aljohani AKB, El-Morsy A, Al Haidari RA, Alamri MAA, Maqnas YA, Alsibaee AM, Mostafa MAH, Keshek DE, Mohamed MG, Bayoumi SAL, Salama SA, El-Adl K. Synthesis and characterization of sodium alginate/poly(N-vinylpyrrolidone) nano-carrier loaded with rebaudioside A and/or stevioside for anticancer drug delivery. Int J Biol Macromol 2025; 316:144778. [PMID: 40449772 DOI: 10.1016/j.ijbiomac.2025.144778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 05/20/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
Stevioside and rebaudioside A revealed anticancer effects against diversity of cancers, such as colon, breast and liver cancers. Rebaudioside A can trigger apoptosis in cancer cells via activation of caspase-dependent pathway. In this study sodium alginate/poly(N-vinylpyrrolidone) as nano-carriers loaded with natural products rebaudioside A (R) and/or stevioside (S) were assessed for anticancer activities. The nanogel of structure R improved cytotoxicity against MCF-7, HepG2, HCT116 and A549 cancers by 60.29 %, 53.45 %, 72.86 % and 62.13 %, correspondingly. Additionally, the nanogel of structure S improved cytotoxicity against MCF-7, HepG2, HCT116 and A549 cancers by 63.96 %, 53.41 %, 70.59 % and 52.88 %, respectively. Furthermore, the nanogel for mixture of R/S improved cytotoxicity against MCF-7, HepG2, HCT116 and A549 cancers by 78.86 %, 54.75 %, 74.10 % and 56.53 % correspondingly. Also, cytotoxic activities of structures R, S and R/S and their nanogels exhibited low toxicity on VERO cells with IC50 = 30.90-46.50 μM and high selectivity against cancer cells. Moreover, R/S (nanogel), R (nanogel) and S (nanogel) demonstrated the uppermost binding affinities with DNA at reduced IC50 values of 31.50, 32.60, and 33.90 μM, respectively. In addition, they inhibited Topo-II activity with remarkably low IC50 value of 0.95, 1.00, and 1.10 μM, respectively.
Collapse
Affiliation(s)
- Shaymaa M M Mohamed
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt; Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Ahmad K B Aljohani
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Ahmed El-Morsy
- Pharmaceutical Chemistry Department, College of Pharmacy, The Islamic University, Najaf, Iraq
| | - Rwaida A Al Haidari
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | | | - Yahya A Maqnas
- College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Aishah M Alsibaee
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Mahmoud A H Mostafa
- Pharmacognosy and Pharmaceutical Chemistry Department, College of Pharmacy, Taibah University, Medina, Saudi Arabia; Department of Pharmacognosy, Faculty of Pharmacy, Al Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Doaa E Keshek
- Department of Biology, Al-Jumum College University, Umm Al-Qura University, P.O Box7388, Makkah 21955, Saudi Arabia; Agriculture Genetic Engineering Research Institute (AGERI), Agriculture Research Centre, Giza, Egypt
| | - May G Mohamed
- Department of Pharmacy Practice, College of Pharmacy, Gulf Medical University, United Arab Emirates
| | - Soad A L Bayoumi
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Khaled El-Adl
- Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt; Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City 11884, Cairo, Egypt.
| |
Collapse
|
11
|
Gupta N, Saha S. Polymer-Based Designer Particles as Drug Carriers: Strategies to Construct and Modify. ACS APPLIED BIO MATERIALS 2025. [PMID: 40405643 DOI: 10.1021/acsabm.5c00041] [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: 05/24/2025]
Abstract
Biological barriers present remarkable challenges for therapeutics delivery, requiring an advanced drug delivery system that can navigate through the complex physiological environment. Polymeric particles provide remarkable versatility due to their adaptable physiochemical properties, facilitating new designs that address complex delivery issues. This review focuses on recent advancements in the morphology of polymeric particles that emulate biological barriers to improve drug efficacy. It includes how structural engineering─such as designing rod-shaped particles for improved cellular uptake, red-blood-cell-shaped particles for prolonged circulation, worm-shaped carriers for improved tissue penetration, and multicompartmental systems for providing combination therapies─profoundly alters drug delivery capabilities. These designer particles exhibit enhanced target specificity, controlled release kinetics, and improved therapeutic outcomes relative to traditional spherical carriers. This particular review also emphasizes how a combination of polymer chemistry and fabrication methods facilitates achieving these advanced structures, while highlighting ongoing challenges in scale-up, reproducibility, and clinical translations. Through the analysis of structure-functional property correlations in various biomimetic designs, we have also attempted to provide insight into future advancements in polymeric delivery systems that have the potential to transform treatment strategies for complicated diseases via shape-directed biological interactions for better therapeutic outcomes.
Collapse
Affiliation(s)
- Nidhi Gupta
- Department of Materials Science and Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- International College of Semiconductor Technology, National Yang-Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Sampa Saha
- Department of Materials Science and Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| |
Collapse
|
12
|
Sun W, Jang MS, Zhan S, Liu C, Sheng L, Lee JH, Fu Y, Yang HY. Tumor-targeting and redox-responsive photo-cross-linked nanogel derived from multifunctional hyaluronic acid-lipoic acid conjugates for enhanced in vivo protein delivery. Int J Biol Macromol 2025; 314:144444. [PMID: 40403518 DOI: 10.1016/j.ijbiomac.2025.144444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 05/14/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
The fabrication of a secure and efficacious nanosystem for intracellular protein delivery is greatly desired, which relies on coordination of the interactions among loading ability, systemic stability, precise tumor targeting, successful endo-lysosomal evasion, and on-demand release characteristics. Herein, we constructed tumor-targeting and redox-responsive photo-crosslinkable nanogels (TRNGs) via UV light-induced ring-opening polymerization (ROP) of lipoic acid moieties incorporated in the side chain of methoxy poly (ethylene glycol) and diethylenetriamine-modified hyaluronic acid (HA-g-mPEG/Deta-c-LA) to create disulfide cross-linked core for the in vivo delivery of cytochrome c (CC). The TRNGs had satisfactory stability for 48 h in physiological environments and high CC encapsulation efficiency via multi-physical interactions. In vivo and in vitro fluorescence imaging proved the preferential accumulation of CC-loaded TRNGs in tumor tissues of human lung tumor-bearing mice and these nanoparticles were efficiently taken up into the CD44-positive A549 cells through CD44-mediated endocytosis compared to CD44-negative HepG2 cells. In addition, the nanoparticles underwent swift exocytosis from the endo-lysosomal compartment, thus promoting the liberation of CC within a reducing intracellular environment. The in vitro therapeutic outcomes proved that empty TRNGs presented excellent biocompatibility and minimal cytotoxicity, whereas CC-loaded TRNGs demonstrated a superior capacity to kill A549 cells compared to free CC and exhibited low effect on CD44-negative HepG2 cells. Moreover, CC-loaded TRNGs also had enhanced antitumor activity without eliciting any adverse effects. Our study highlighted the potential of TRNGs as a novel nanoplatform for the treatment of protein-based cancers.
Collapse
Affiliation(s)
- Wei Sun
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China
| | - Moon-Sun Jang
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Siqi Zhan
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China
| | - Changling Liu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China
| | - Li Sheng
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China
| | - Jung Hee Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea.
| | - Yan Fu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China.
| | - Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, PR China.
| |
Collapse
|
13
|
Austria ES, Akhavan B. Polymeric nanoparticle synthesis for biomedical applications: advancing from wet chemistry methods to dry plasma technologies. NANOSCALE 2025. [PMID: 40391562 DOI: 10.1039/d5nr00436e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
Nanotechnology has introduced a transformative leap in healthcare over recent decades, particularly through nanoparticle-based drug delivery systems. Among these, polymeric nanoparticles (NPs) have gained significant attention due to their tuneable physicochemical properties for overcoming biological barriers. Their surfaces can be engineered with chemical functional groups and biomolecules for a wide range of biomedical applications, ranging from drug delivery to diagnostics. However, despite these advancements, the clinical translation and large-scale commercialization of polymeric NPs face significant challenges. This review uncovers these challenges by examining the interplay between structural design and payload interaction mode. It provides a critical evaluation of the current synthesis methods, beginning with conventional wet chemical techniques, and progressing to emerging dry plasma technologies, such as plasma polymerization. Special attention is given to plasma polymerized nanoparticles (PPNs), highlighting their potential as paradigm-shifting platforms for biomedical applications while identifying key areas for improvement. The review concludes with a forward-looking discussion on strategies to address key challenges, such as achieving regulatory approval and advancing clinical translation of polymeric NP-based therapies, offering unprecedented opportunities for next-generation nanomedicine.
Collapse
Affiliation(s)
- Elmer S Austria
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, NSW 2006, Australia.
- Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Behnam Akhavan
- School of Biomedical Engineering, Faculty of Engineering, University of Sydney, Sydney, NSW 2006, Australia.
- Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
- School of Engineering, College of Engineering, Science and Environment, University of Newcastle, Callaghan, NSW 2308, Australia
- Hunter Medical Research Institute (HMRI), Precision Medicine Program, New Lambton Heights, NSW 2305, Australia
| |
Collapse
|
14
|
Sahane P, Puri N, Khairnar P, Phatale V, Shukla S, Priyadarshinee A, Srivastava S. Harnessing Folate Receptors: A Comprehensive Review on the Applications of Folate-Adorned Nanocarriers for the Management of Melanoma. ACS APPLIED BIO MATERIALS 2025; 8:3623-3656. [PMID: 40275606 DOI: 10.1021/acsabm.5c00077] [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: 04/26/2025]
Abstract
The advancement in exclusively tailored therapeutic delivery systems has escalated a great deal of interest in targeted delivery to augment therapeutic efficacy and to lessen adverse effects. The targeted delivery approach promisingly helps to surmount the unmet clinical needs of conventional therapies, including chemoresistance, limited penetration, and side effects. In the case of melanoma, various receptors were overexpressed on the tumor site, among which folate receptor (FR) targeting is considered to be a progressive approach for managing melanoma. FRs are the macromolecules of the glycosyl phosphatidylinositol-attached protein that possess globular assembly with a greater affinity toward specific ligands. So, the functional ligands can be utilized to design targeted nanocarriers (NCs) that can effectively bind to overexpressed FRs. Hence, folate-adorned NCs (FNCs) offer various benefits such as site-specific targeting, cargo protection, and minimizing toxicity. This review focuses on the insights and implications of FRs, targeting FRs, and mechanisms, challenges, and advantages of FNCs. Further, the applications of various FNCs, such as liposomes, polymeric NCs, albumin nanoparticles, inorganic NCs, liquid crystalline nanoparticles, and nanogels, have been elaborated for melanoma therapy. Likewise, the potential of FNCs in immunotherapy, photodynamic therapy, chemotherapy, gene therapy, photothermal therapy, and tumor imaging has been exhaustively discussed. Furthermore, translational hurdles and potential solutions are discussed in detail. The present review is expected to give thoughtful ideas to researchers, industry stakeholders, and formulation scientists for the efficacious development of FNCs.
Collapse
Affiliation(s)
- Prajakta Sahane
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Niharika Puri
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Shalini Shukla
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Abhipsa Priyadarshinee
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, Telangana, India
| |
Collapse
|
15
|
Aldhubiab B, Almuqbil RM, Nair AB. Harnessing the Power of Nanocarriers to Exploit the Tumor Microenvironment for Enhanced Cancer Therapy. Pharmaceuticals (Basel) 2025; 18:746. [PMID: 40430563 PMCID: PMC12115050 DOI: 10.3390/ph18050746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
The tumor microenvironment (TME) has a major role in malignancy and its complex nature can mediate tumor survival, metastasis, immune evasion, and drug resistance. Thus, reprogramming or regulating the immunosuppressive TME has a significant contribution to make in cancer therapy. Targeting TME with nanocarriers (NCs) has been widely used to directly deliver anticancer drugs to control TME, which has revealed auspicious outcomes. TME can be reprogrammed by using a range of NCs to regulate immunosuppressive factors and activate immunostimulatory cells. Moreover, TME can be ameliorated via regulating the redox environment, oxygen content, and pH value of the tumor site. NCs have the capacity to provide site-specific delivery of therapeutic agents, controlled release, enhanced solubility and stability, decreased toxicities, and enhanced pharmacokinetics as well as biodistribution. Numerous NCs have demonstrated their potential by inducing distinct anticancer mechanisms by delivering a range of anticancer drugs in various preclinical studies, including metal NCs, liposomal NCs, solid lipid NCs, micelles, nanoemulsions, polymer-based NCs, dendrimers, nanoclays, nanocrystals, and many more. Some of them have already received US Food and Drug Administration approval, and some have entered different clinical phases. However, there are several challenges in NC-mediated TME targeting, including scale-up of NC-based cancer therapy, rapid clearance of NCs by the mononuclear phagocyte system, and TME heterogeneity. In order to harness the full potential of NCs in tumor treatment, there are several factors that need to be carefully studied, including optimization of drug loading into NCs, NC-associated immunogenicity, and biocompatibility for the successful translation of NC-based anticancer therapies into clinical practice. In this review, a range of NCs and their applications in drug delivery to remodel TME for cancer therapy are extensively discussed. Moreover, findings from numerous preclinical and clinical studies with these NCs are also highlighted.
Collapse
Affiliation(s)
- Bandar Aldhubiab
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (R.M.A.); (A.B.N.)
| | | | | |
Collapse
|
16
|
Mittal G, A P, Dhali A, Prasad R, S Y, Nurani KM, Găman MA. Plant extracts with antioxidant and hepatoprotective benefits for liver health: A bibliometric analysis of drug delivery systems. World J Gastroenterol 2025; 31:105836. [DOI: 10.3748/wjg.v31.i18.105836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/07/2025] [Accepted: 04/21/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND The rising global burden of liver diseases, such as non-alcoholic fatty liver disease and liver fibrosis, has necessitated innovative therapeutic approaches. Plant-based therapies, recognized for their anti-inflammatory and antioxidant properties, have shown promising effects. However, poor bioavailability limits their clinical application.
AIM To map global research trends, key contributors, and emerging themes in plant-based therapies combined with advanced drug delivery systems for liver health.
METHODS Using the Scopus database, 645 documents were retrieved and analyzed using bibliometric tools Biblioshiny and VOSviewer. Analysis focused on publication trends, geographical contributions, and advancements in drug delivery technologies, including nanoparticles, liposomes, and polymeric micelles. Metrics such as publication growth rate, authorship collaboration, and thematic clustering were assessed.
RESULTS The dataset spans 43 years (1981-2024), with an annual growth rate of 11.09% in the number of publications. Research output is dominated by China (33%), followed by the United States (24%) and India (18%). Collaborative studies accounted for 24.34% of publications, with an average of 5.81 co-authors per document. Key innovations include nanoparticle encapsulation of curcumin and silymarin, improving bioavailability by up to 85%. Highly cited studies demonstrated the antioxidant, anti-inflammatory, and anti-fibrotic properties of these compounds. For instance, curcumin nanoparticles showed a 70% improvement in solubility, and silymarin liposomal formulations enhanced therapeutic efficiency by 62%. Thematic analysis revealed a transition from basic clinical observations to molecular and pharmacokinetic research, with a focus on oxidative stress mitigation and hepatoprotection.
CONCLUSION This study highlights the growing synergy between plant-based therapies and advanced drug delivery systems, with significant contributions from Asian and Western countries. Future efforts should prioritize clinical trials, standardization of plant extract formulations, and interdisciplinary approaches to maximize therapeutic outcomes. The findings provide a foundation for integrating plant-derived compounds into evidence-based hepatological therapies, addressing critical challenges in bioavailability and safety.
Collapse
Affiliation(s)
- Gaurav Mittal
- MBBS Final Year StudentMahatma Gandhi Institute of Medical Sciences, Maharashtra 442102, India
| | - Prashanth A
- Department of Physiology, Mahatma Gandhi Institute of Medical Sciences, Maharashtra 442102, India
| | - Arkadeep Dhali
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield S5 7AU, United Kingdom
| | - Roshan Prasad
- Department of Medicine, Datta Meghe Institute of Higher Education and Research, Sawangi 442107, India
| | - Yogesh S
- Department of Medicine, Madras Medical College, Chennai 600003, India
| | | | - Mihnea-Alexandru Găman
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Hematology, Centre of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest 022328, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, Bucharest 010001, Romania
| |
Collapse
|
17
|
Dai Y, Yang L, Cao G, Mo L, Yang C, Zhu Y, Guo Y, Hong Y, Xu H, Lu S, Du S, He J. Combination therapy and drug co-delivery systems for atherosclerosis. J Control Release 2025; 381:113543. [PMID: 39986476 DOI: 10.1016/j.jconrel.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/25/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of plaque within the arteries. Despite advances in therapeutic strategies including anti-inflammatory, antioxidant, and lipid metabolism modulation treatments over the past two decades, the treatment of atherosclerosis remains challenging, as arterial damage is the result of interconnected pathological factors. Therefore, current monotherapies often fail to address the complex nature of this disease, leading to insufficient therapeutic outcomes. This review addressed this paucity of effective treatment options by comprehensively exploring the potential for combination therapies and advanced drug co-delivery systems for the treatment of atherosclerosis. We investigated the pathological features of and risk factors for atherosclerosis, underscoring the importance of drug combination therapies for the treatment of atherosclerotic diseases. We discuss herein mathematical models for quantifying the efficacy of the combination therapies and provide a systematic summary of drug combinations for the treatment of atherosclerosis. We also provide a detailed review of the latest advances in nanoparticle-based drug co-delivery systems for the treatment of atherosclerosis, focusing on the design of carriers with high biocompatibility and efficacy. By exploring the possibilities and challenges inherent to this approach, we aim to highlight cutting-edge technologies that can foster the development of innovative strategies, optimize drug co-administration, improve treatment outcomes, and reduce the burden of atherosclerosis-related morbidity and mortality on the healthcare system.
Collapse
Affiliation(s)
- Yingxuan Dai
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Li Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Guosheng Cao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China
| | - Liqing Mo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Can Yang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yuxi Zhu
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Department of Pediatrics, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, OH 44106, USA
| | - Yujie Guo
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Yi Hong
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Hanlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China
| | - Shan Lu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| | - Shi Du
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210, USA; Division of Pharmaceutics and Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210, USA.
| | - Jianhua He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, PR China; Hubei Shizhen Laboratory, Wuhan 430065, PR China; Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, PR China.
| |
Collapse
|
18
|
Hu L, Ma H, Yang C, Wang K, Ji X, Xu T, Xu H, Zhao Y, Zhen Y. Environmental responsive dual-drug synergistic and dual-targeted polymer micelles based on chondroitin sulfate for treatment of breast cancer. Int J Biol Macromol 2025; 311:144083. [PMID: 40348243 DOI: 10.1016/j.ijbiomac.2025.144083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 04/23/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Targeted drug delivery strategy can accurately deliver multiple drugs to tumor cells by biocompatible materials, enhance the therapeutic effect and weaken adverse side effects. Chondroitin sulfate (CS) is a biodegradable material which has dual-targeting ability to CD44 receptors and Golgi apparatus. Flurbiprofen is a COX-2 inhibitor that can be used as an anticancer adjuvant in combination with docetaxel to enhance the antitumor effect. Herein, docetaxel and flurbiprofen were coupled to CS via disulfide bond and ester bond respectively to prepare polymer conjugates chondroitin sulfate-docetaxel (C-ss-D) and chondroitin sulfate-flurbiprofen (C-F). Then the reduction-sensitive micelles C-ss-D and the pH-sensitive micelles C-F were constructed by self-assembly respectively, and the dual-drug micelles C-F/C-ss-D were further prepared. C-F/C-ss-D displayed uniform spherical shape, negative surface charge, and achieved controlled drug release in slightly acidic and reductive tumor microenvironment. C-F/C-ss-D exhibited remarkable targeting ability towards tumor cells and Golgi apparatus, demonstrated potent cytotoxic effect on MCF-7 cells, and induced apoptosis by regulating the expression of COX-2 and apoptosis-related proteins. C-F/C-ss-D effectively inhibited tumor growth in MCF-7 xenograft mice with low toxicity to blood and major organs. Therefore, the environmental responsive dual-drug synergistic and dual-targeted micelles based on CS have great potential for the treatment of breast cancer.
Collapse
Affiliation(s)
- Litao Hu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Huiling Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Chunpeng Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Kang Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Xu Ji
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Tingxue Xu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Hong Xu
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
19
|
Sun H, Zhao J, Liu B, Gou J, He H, Zhang Y, Yin T, Jin X, Tang X. Integrity of polymeric micelles regulates bioavailability of cyclosporine A: A FRET-based analysis during oral delivery. J Control Release 2025; 383:113831. [PMID: 40349783 DOI: 10.1016/j.jconrel.2025.113831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 05/06/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
This study comprehensively investigated the effect of structural integrity on the oral delivery efficacy of mPEG-b-PCL polymeric micelles (PMs) using fluorescence resonance energy transfer (FRET) technology. During mucus penetration, the integrity loss of PMs resulted from the "hydration effect" of mucins and the "solubilization effect" of bile salts. Increasing surface PEG density or decreasing particle size <50 nm resisted the "hydration effect", while shortening PCL chain or increasing aggregation number alleviated the "solubilization effect". Fluorescence colocalization studies demonstrated that low surface PEG density and small particle size enhanced the cellular uptake of PMs, but high surface PEG density preserved the intracellular integrity of PMs. Approximately 30 %-60 % of endocytosed PMs were expelled within 4 h, correlating with their cellular uptake efficiency. Transcellular transport results indicated that 5 %-25 % of PMs underwent transcytosis within 4 h, 0.5 %-10 % of initial PMs remained intact. In situ intestine perfusion data confirmed the consistency between the in vitro findings and the actual performance of PMs in vivo. Two types of PMs were selected to encapsulate cyclosporine A (CyA) based on their enhanced integrity and transcytosis efficiency. The PM with a short amphiphilic block and loose structure exhibited comparable in vitro release and in vivo performance to Neoral®, with a Cmax of 1356.28 ± 170.01 ng/mL, a Tmax of 2 h, and an AUC0-t of 16,408.92 ± 1166.78 ng/mL*h. However, the PM with a long amphiphilic block and ordered core PCL arrangement exhibited sustained release of CyA both in vitro and in vivo, decreasing CyA accumulation in major organs and improving the oral bioavailability of CyA, with a Cmax of 757.07 ± 66.19 ng/mL, a Tmax of 9.33 ± 2.31 h, and an AUC0-t of 21,938.44 ± 2183.59 ng/mL*h.
Collapse
Affiliation(s)
- Haihan Sun
- School of Pharmaceutical Sciences, Jilin University, No. 1266, Fujin Road, Changchun 130021, China
| | - Jiansong Zhao
- School of Pharmaceutical Sciences, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Boyuan Liu
- School of Pharmaceutical Sciences, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Jingxin Gou
- School of Pharmaceutical Sciences, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Haibing He
- School of Pharmaceutical Sciences, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Yu Zhang
- School of Pharmaceutical Sciences, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Xiangqun Jin
- School of Pharmaceutical Sciences, Jilin University, No. 1266, Fujin Road, Changchun 130021, China.
| | - Xing Tang
- School of Pharmaceutical Sciences, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
20
|
Motta MA, Martin-Saldaña S, Beloqui A, Calderón M, Larrañaga A. Polypeptide-based multilayer capsules with anti-inflammatory properties: exploring different strategies to incorporate hydrophobic drugs. J Mater Chem B 2025; 13:5297-5314. [PMID: 40207430 DOI: 10.1039/d4tb01906g] [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: 04/11/2025]
Abstract
More than 90% of drug candidates used in the drug development pipeline and about 40% of drugs on the market are poorly soluble in water based on the definition of the biopharmaceutical classification system. The advent of drug delivery approaches has represented a striking tool to overcome the challenges associated with the use of hydrophobic drugs, such as their low bioavailability and off-target effects. Drug carrier formulations composed of biodegradable and biocompatible polymers, such as polypeptides, have been explored as platforms to host poorly water-soluble drugs to prolong drug circulation, enhance their safety, reduce their immunogenicity, and promote their controlled release. In this work, we evaluated three strategies-co-precipitation, post-encapsulation, and conjugation-to incorporate a hydrophobic model drug, i.e., curcumin (CUR), into biodegradable multilayer capsules fabricated via a layer-by-layer (LbL) approach. Poly(L-lysine) (PLys) and poly(L-glutamic acid) (PGlu) were adopted as building blocks and alternately assembled onto calcium carbonate (CaCO3) microparticles to build a polypeptide-multilayer membrane, which acted as a barrier to control the release of the drug. The application of our three formulations in in vitro inflammatory models of THP-1 derived human macrophages and murine microglia showed a reduction of the inflammation with the suppression of three pivotal pro-inflammatory cytokines (i.e., interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α). Moreover, the intracellular release of CUR detected upon uptake studies on activated microglia suggested that our systems could represent a potential therapeutic approach to reduce acute neuroinflammation and modulate microglia phenotype.
Collapse
Affiliation(s)
- Maria Angela Motta
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, 48013 Bilbao, Spain.
| | - Sergio Martin-Saldaña
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
| | - Ana Beloqui
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Marcelo Calderón
- POLYMAT, Applied Chemistry Department, Faculty of Chemistry, University of the Basque Country UPV/EHU, Paseo Manuel de Lardizabal 3, 20018 Donostia-San Sebastián, Spain.
- IKERBASQUE, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Aitor Larrañaga
- Department of Mining-Metallurgy Engineering and Materials Science, POLYMAT, Bilbao School of Engineering, University of the Basque Country (UPV/EHU), Plaza Torres Quevedo 1, 48013 Bilbao, Spain.
| |
Collapse
|
21
|
Uddin MJ, Lo JHJ, Gupta MK, Werfel TA, Asaduzzaman A, Oltman CG, Gbur EF, Mohyuddin MT, Nazmin F, Rahman MS, Jashim A, Crews BC, Kingsley PJ, Klendworth JE, Marnett LJ, Duvall CL, Cook RS. Polymeric Nanoparticles Enable Targeted Visualization of Drug Delivery in Breast Cancer. Mol Pharm 2025; 22:2392-2401. [PMID: 40257460 PMCID: PMC12056698 DOI: 10.1021/acs.molpharmaceut.4c00695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/22/2025]
Abstract
We report the coencapsulation of fluorocoxib Q (FQ) and chemocoxib A (CA) in micellar nanoparticles (FQ-CA-NPs) of a new PPS135-b-POEGA17 diblock polymer, which exhibited a hydrodynamic diameter of 109.2 ± 4.1 nm and a zeta potential (ζ) of -1.59 ± 0.3 mV. The uptake of FQ-CA-NPs by 4T1 mouse mammary cancer cells and intracellular cargo release were assessed by fluorescence microscopy that resulted in increased fluorescence in 4T1 cells compared to cells pretreated with celecoxib. The viability of primary human mammary epithelial cells (HMECs) or 4T1 mouse mammary carcinoma cells treated with FQ-CA-NPs were assessed, which showed decreased growth of 4T1 breast cancer cells but showed no effect on the growth of primary human mammary epithelial cells (HMECs). Intravenous dosing of FQ-CA-NPs in mice enabled ROS-induced cargo (FQ and CA) release and fluorescence activation of FQ and resulted in increased fluorescence in breast tumors compared to the tumors of animals pretreated with tempol or celecoxib, and minimum fluorescence was detected in the tumors of animals treated with nothing or empty-NPs. In addition, tumor tissues from treated animals were analyzed ex vivo by liquid chromatography-mass spectrometry (LC-MS)/MS, and identified increased levels of cargo delivery and retention in the tumor compared to tempol- or celecoxib-pretreated animal tumors. These in vivo and ex vivo results confirmed the targeted delivery of loaded NPs followed by ROS-mediated cargo release and fluorescence activation for targeted visualization of drug delivery in breast tumors and CA-induced therapeutic effect in an in vivo tumor growth inhibition assay and an ex vivo hematoxylin and eosin (H&E) staining of tumor tissues. Thus, coencapsulation of FQ and CA into polymeric micellar nanoparticles (FQ-CA-NPs) enabled their ROS-sensitive release followed by fluorescence activation and COX-2-dependent tumor targeting and retention in the visualization of CA delivery in solid breast tumors.
Collapse
Affiliation(s)
- Md. Jashim Uddin
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Justin Han-Je Lo
- Department
of Medicine, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Mukesh K. Gupta
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Thomas A. Werfel
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Abu Asaduzzaman
- Departments
of Electrical and Computer Engineering, Wichita State University School of Engineering, Wichita, Kansas 67260, United States
| | - Connor G. Oltman
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Eva F. Gbur
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Mohammed T. Mohyuddin
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Farhana Nazmin
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Md. Saidur Rahman
- Department
of Medicine, Vanderbilt University Medical
Center, Nashville, Tennessee 37232, United States
| | - Ahan Jashim
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Brenda C. Crews
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Philip J. Kingsley
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Jamie E. Klendworth
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Lawrence J. Marnett
- Departments
of Biochemistry, Chemistry and Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Craig L. Duvall
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| | - Rebecca S. Cook
- Department
of Biomedical Engineering, Vanderbilt University
School of Engineering, Nashville, Tennessee 37232, United States
| |
Collapse
|
22
|
Madheswaran T, Chellappan DK, Lye FSN, Dua K. Recent advances in the use of liquid crystalline nanoparticles for non-small cell lung cancer treatment. Expert Opin Drug Deliv 2025; 22:615-627. [PMID: 40022612 DOI: 10.1080/17425247.2025.2474693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/24/2025] [Accepted: 02/27/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) continues to pose a considerable health challenge with few therapeutic alternatives. Liquid crystalline nanoparticles (LCN) are nanostructured drug delivery systems made of lipid-based amphiphilic materials that self-assemble into crystalline phases in aqueous environments. LCN have become a promising way to treat NSCLC owing to their specific properties that make them useful for targeted delivery and controlled drug release. AREAS COVERED The review provides a brief overview of the use of LCN in the treatment of NSCLC. It explores their composition, fabrication methods, and characterization processes. The article further addresses several nanoparticle-based approaches for the treatment of NSCLC. Ultimately, it underscores the promise of LCNs as a promising drug delivery system for NSCLC and discusses the obstacles and outlook in this field. EXPERT OPINION LCN represents a promising frontier in the treatment of NSCLC, offering several specific advantages over conventional therapies. Utilizing their intrinsic self-assembly characteristics, LCN provides meticulous control over drug encapsulation, release kinetics, and cellular absorption, which are crucial for improving therapy success. LCN also has the capability for co-delivery of various drugs, facilitating synergistic therapeutic benefits and addressing multidrug resistance, a prevalent issue in NSCLC treatment.
Collapse
Affiliation(s)
- Thiagarajan Madheswaran
- Department of Pharmaceutical Technology, School of Pharmacy, IMU University, Kuala Lumpur, Malaysia
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research Development and Innovation, IMU University, Kuala Lumpur, Malaysia
| | - Dinesh Kumar Chellappan
- Centre for Bioactive Molecules and Drug Delivery, Institute for Research Development and Innovation, IMU University, Kuala Lumpur, Malaysia
- Department of Life Sciences, School of Pharmacy, IMU University, Kuala Lumpur, Malaysia
| | - Fiona Sze Nee Lye
- School of Postgraduate Studies, IMU University, Kuala Lumpur, Selangor, Malaysia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, NSW, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, Australia
- Woolcock Institute of Medical Research, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
23
|
Yang C, Ma H, Liang Z, Zhuang Y, Hu L, Zhang K, Huang L, Li M, Zhang S, Zhen Y. Cyclic RGD modified dextran-quercetin polymer micelles for targeted therapy of breast cancer. Int J Biol Macromol 2025; 308:142272. [PMID: 40118409 DOI: 10.1016/j.ijbiomac.2025.142272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Quercetin is a natural flavonoid found in many plants which has various pharmacological activities including antitumor effect. However, the poor water solubility and bioavailability limit the potential benefits of quercetin for patients. Thus, modifying quercetin structure and developing actively targeted drug delivery systems are extremely important for tumor precision therapy. Herein, polymer-drug conjugates dextran-quercetin (D-Q) and cRGD-dextran (R-D) were synthesized by grafting quercetin and polypeptide cRGDfk (Arg-Gly-Asp-(D-Phe)-Lys) to dextran. Then cRGD-modified dextran-quercetin polymer micelles (R-D-Q) were constructed by self-assembling of D-Q and R-D. R-D-Q micelles possessed appropriate particle size (133.4 nm), nearly neutral potential (8.14 mV) and excellent drug-loading efficiency (13.1 %) and achieved higher cytotoxicity, apoptosis induction and penetration to human breast cancer MCF-7 cells than the micelles unmodified with cRGD, which were ascribed to cRGD-integrin mediated transcytosis. R-D-Q micelles effectively suppressed tumor growth in tumor-bearing mice by delivering more quercetin throughout the tumor tissue. And R-D-Q micelles could promote the apoptosis of tumor cells by activating p38 and JNK signal pathways and suppressing ERK signal pathway. In addition, R-D-Q micelles had no damage to normal tissues of mice at therapeutic dose. These results indicate promising prospects for R-D-Q micelles as an effective drug delivery system against tumor.
Collapse
Affiliation(s)
- Chunpeng Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Huiling Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ze Liang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ying Zhuang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Litao Hu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Kexin Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Leixiao Huang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Min Li
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, China; Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
24
|
Gothwal A, Muolokwu CE, Chaulagain B, Mahanta AK, Singh J. Intranasal delivery of pApoE2 via penetratin-mannose multi-functionalized chitosan polymeric micelles to the brain: Reduced total tau and phosphorylated tau burden in transgenic Alzheimer's mouse model. Int J Biol Macromol 2025; 310:143542. [PMID: 40294672 DOI: 10.1016/j.ijbiomac.2025.143542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The phosphorylated tau accumulation is a classical pathological hallmark of future cognitive decline and a cause of neuronal death in Alzheimer's disease (AD). In this study, we developed multi-functionalized chitosan (CS) based polymeric micelles to effectively deliver pApoE2 via intranasal administration to the brain. The CS was modified with caproic acid (CA), cell-penetrating peptide penetratin (PEN), and GLUT-1 transporter ligand mannose (MAN) for selective and enhanced delivery to the brain. The multi-functionalized Cap-g-CS-PEN-MAN polymeric micelles were ≤200 nm in size, cationic in charge, and uniformly distributed (PDI ≤ 0.3). The multi-functionalized polymeric micelles did not exhibit toxicity against bEnd.3 cells and erythrocytes up to polymer concentrations of 500 μg/mL. The Cap-g-CS-PEN-MAN /pDNA polyplex was stable against a DNase rich environment. The Cap-g-CS-PEN-MAN/pAPoE2 polyplex demonstrated elevated expression of ApoE in primary astrocytes and neurons, 9.47 ± 2.13 and 5.67 ± 2.69 ng/mg of protein, respectively. The therapeutic efficacy of the Cap-g-CS-PEN-MAN/pApoE2 polyplex was analyzed against the PS19 tauopathy mice model. Total tau burden was significantly (p ≤ 0.05) reduced by 4.09 ± 1.4 ng/mg of protein in Cap-g-CS-PEN-MAN/pApoE2 polyplex administered mice over the other groups. Phosphorylated tau pT181 level was also significantly (p ≤ 0.05) reduced in Cap-g-CS-PEN-MAN/pApoE2 polyplex administered mice over saline, pApoE2 and Cap-g-CS/pApoE2 treated groups.
Collapse
Affiliation(s)
- Avinash Gothwal
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Chinenye Edith Muolokwu
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Bivek Chaulagain
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Arun Kumar Mahanta
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, ND 58108-6050, USA.
| |
Collapse
|
25
|
Chen P, Ren L, Guo Y, Sun Y. Boosting antitumor immunity in breast cancers: Potential of adjuvants, drugs, and nanocarriers. Int Rev Immunol 2025; 44:141-164. [PMID: 39611269 DOI: 10.1080/08830185.2024.2432499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/05/2024] [Accepted: 11/17/2024] [Indexed: 11/30/2024]
Abstract
Despite advancements in breast cancer treatment, therapeutic resistance, and tumor recurrence continue to pose formidable challenges. Therefore, a deep knowledge of the intricate interplay between the tumor and the immune system is necessary. In the pursuit of combating breast cancer, the awakening of antitumor immunity has been proposed as a compelling avenue. Tumor stroma in breast cancers contains multiple stromal and immune cells that impact the resistance to therapy and also the expansion of malignant cells. Activating or repressing these stromal and immune cells, as well as their secretions can be proposed for exhausting resistance mechanisms and repressing tumor growth. NK cells and T lymphocytes are the prominent components of breast tumor immunity that can be triggered by adjuvants for eradicating malignant cells. However, stromal cells like endothelial and fibroblast cells, as well as some immune suppressive cells, consisting of premature myeloid cells, and some subsets of macrophages and CD4+ T lymphocytes, can dampen antitumor immunity in favor of breast tumor growth and therapy resistance. This review article aims to research the prospect of harnessing the power of drugs, adjuvants, and nanoparticles in awakening the immune reactions against breast malignant cells. By investigating the immunomodulatory properties of pharmacological agents and the synergistic effects of adjuvants, this review seeks to uncover the mechanisms through which antitumor immunity can be triggered. Moreover, the current review delineates the challenges and opportunities in the translational journey from bench to bedside.
Collapse
Affiliation(s)
- Ping Chen
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Lei Ren
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Youwei Guo
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| | - Yan Sun
- Pharmacy Department, Zibo Central Hospital, Zibo, Shandong, China
| |
Collapse
|
26
|
Sun Z, Fu H, Zhang R, Wang H, Shen S, Zhao C, Li X, Sun Y, Li Y, Li Y. Advances in chemically modified HSA as a multifunctional carrier for transforming cancer therapy regimens. Int J Biol Macromol 2025; 305:141373. [PMID: 39988174 DOI: 10.1016/j.ijbiomac.2025.141373] [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: 12/11/2024] [Revised: 02/04/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Human serum albumin (HSA) is a versatile, biodegradable, biocompatible, non-toxic, and non-immunogenic protein nanocarrier, making it an ideal platform for developing advanced drug delivery systems. These properties have garnered significant attention in utilizing HSA nanoparticles for the safe and efficient delivery of chemotherapeutic agents. HSA-based nanoparticles can be surface-modified with various ligands to enable tumor-targeted drug delivery, enhancing therapeutic specificity and efficacy. Furthermore, the multifunctionality of HSA nanoparticles offers promising strategies to overcome challenges in cancer therapy, including poor bioavailability, off-target toxicity, and drug resistance. This review highlights the structural features of HSA, explores its diverse modifications to improve drug-binding affinity and targeting ability, and discusses its potential as a multifunctional carrier in oncology. By summarizing the latest advances in HSA modification techniques and applications, this review provides a comprehensive perspective on the future of protein-based drug delivery systems in tumor therapy.
Collapse
Affiliation(s)
- Zheng Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hui Fu
- School of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruixuan Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Hui Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shiyang Shen
- School of Integrated Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chengcheng Zhao
- Experimental Teaching and Practical Training Center, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Xiuyan Li
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150040, China
| | - Yujiao Sun
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yunfei Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Yingpeng Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
27
|
Babanyinah GK, Bhadran A, Polara H, Shah T, Biewer MC, Stefan MC. Fluorescent Poly(ε-Caprolactone)s Micelles for Anticancer Drug Delivery and Bioimaging. Biomacromolecules 2025. [PMID: 40305416 DOI: 10.1021/acs.biomac.5c00262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Despite significant advancements in polymer-based nanomedicine, the clinical translation of biodegradable micellar drug delivery systems is limited due to premature drug release, low drug-loading capacity (DLC), and lack of inherent therapeutic and bioimaging functionalities. To overcome these challenges, we designed a novel poly(ε-caprolactone) (PCL)-based amphiphilic diblock copolymer that possesses inherent anticancer activity, fluorescence imaging capabilities, and multistimuli-responsive drug release. This platform features a fluorescent hydrophilic shell comprising of two triethylene glycol units and a hydrophobic core containing anticancer naphthalene moieties. This unique architecture imparts remarkable properties: the triethylene glycol units confer thermoresponsive behavior for precise drug release and enable intracellular tracking, while the naphthalene pendants enhance DLC (3.7%) through π-π interactions with doxorubicin (DOX). The micelles exhibit a low critical micelle concentration (7.8 × 10-3 g/L), demonstrate strong stability for long storage times, and show significant cytotoxicity against the MDA-MB-231 cell line, highlighting their combined therapeutic efficacy.
Collapse
Affiliation(s)
- Godwin K Babanyinah
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Abhi Bhadran
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Himanshu Polara
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Tejas Shah
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Michael C Biewer
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Mihaela C Stefan
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
28
|
Zhang X, Zhao B, Fu S, Seruya RS, Fanos HE, Petrisor AA, Liu Y, Yang Z, Zhang F. Controlling stimulus sensitivity by tailoring nanoparticle core hydrophobicity. Biomater Sci 2025; 13:2332-2339. [PMID: 40138203 PMCID: PMC11939950 DOI: 10.1039/d5bm00163c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a significant global health challenge, necessitating the development of more effective therapeutic strategies. This work presents a novel glutathione (GSH)-responsive platform designed to enhance the delivery and efficacy of the anticancer drug mertansine (DM1) through the modulation of pendant groups in polycarbonate-drug conjugates. By systematically varying the hydrophobicity of the pendant groups, we investigated their effects on nanostructures, GSH sensitivity, colloidal stability, drug release profiles, and the in vitro anticancer efficacy of these polymeric nanoparticles, revealing that more hydrophobic pendant groups effectively reduce GSH accessibility for the nanoparticle cores, improve colloidal stability, and slow drug release rates. The results underscore the critical importance of polymer structures in optimizing drug delivery systems and offer valuable insights for future research on advanced nanomaterials with enhanced drug delivery for cancer therapies.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Bowen Zhao
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Shiwei Fu
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Ronald S Seruya
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Hannah E Fanos
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Ashley A Petrisor
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Yilin Liu
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Zixin Yang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Fuwu Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- The Dr John T. Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, 1951 NW 7th Ave, Miami, Florida, 33136, USA
| |
Collapse
|
29
|
Nguyen TTL, Duong VA. Advancements in Nanocarrier Systems for Nose-to-Brain Drug Delivery. Pharmaceuticals (Basel) 2025; 18:615. [PMID: 40430435 PMCID: PMC12115159 DOI: 10.3390/ph18050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
In recent decades, nose-to-brain drug delivery has shown effectiveness in treating many central nervous system diseases. Intranasally administered drugs can be delivered to the brain through the olfactory and trigeminal pathways that bypass the blood-brain barrier. However, nose-to-brain drug delivery is challenging due to the inadequate nasal mucosa absorption of drugs and the short retention time of the intranasal formulations. These problems can be minimized through the use of nano-drug delivery systems, such as micelles, polymeric nanoparticles, nanoemulsions, liposomes, solid lipid nanoparticles, and nanostructured lipid carriers. They can enhance the drug's bioavailability in the brain via increases in drug solubility, permeation, and stability. Nose-to-brain nano-drug delivery systems have been evaluated in vivo by a number of research groups. This review aims to provide an overview of nose-to-brain delivery and recent advances in the development of nano-drug delivery systems for delivering drugs from the nose to the brain to improve the treatment of some central nervous system diseases.
Collapse
Affiliation(s)
- Thi-Thao-Linh Nguyen
- Institute of Pharmaceutical Education and Research, Binh Duong University, Thu Dau Mot City 820000, Binh Duong, Vietnam;
| | - Van-An Duong
- The Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
30
|
Pan J, Wang H, Liu Z, Jiang D, Yuan M. Study on the Self-assembly Behavior of Polycaprolactone Star-Shaped Copolymers Based on Dissipative Particle Dynamics. ACS OMEGA 2025; 10:15419-15431. [PMID: 40291000 PMCID: PMC12019736 DOI: 10.1021/acsomega.5c00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
In this study, the pH-responsive self-assembly behavior of polycaprolactone star-shaped block copolymers in aqueous solution was systematically investigated by dissipative particle dynamics. The changes in morphology during the self-assembly process were studied by adjusting the number of polymer star arms, the degree of ionization of the terminal carboxylic acid group, and the concentration of the polymer in the solution. The results show that during the self-assembly process, the star-shaped copolymers undergo a series of structural transformations from spherical micelles to worm-like micelles and then to lamellar micelles as ionization and solution concentration change. When the degree of ionization is high, the electrostatic repulsion is enhanced, resulting in the formation of smaller spherical micelles; in contrast, when the degree of ionization is low, the electrostatic repulsion is weakened, resulting in the formation of larger and more complex worm-like or lamellar structures. With an increase in the number of arms, the self-assembly behavior of the system gradually transitions from complex morphologies (such as lamellar micelles and branched worm-like micelles) to simple morphologies (such as linear worm-like micelles and spherical micelles). This study provides important theoretical references and practical guidance for revealing the regulatory mechanisms of carboxyl ionization, the number of polymer star arms, and solution concentration on the self-assembly behavior of amphiphilic block copolymers.
Collapse
Affiliation(s)
- Jiashu Pan
- Green
Preparation Technology of Biobased Materials National & Local
joint Engineering Research Center, Yunnan
Minzu University, Kunming 650500, China
| | - Haiming Wang
- Affiliated
School of Kunming NSAU Research Institute, Kunming 650500, China
| | - Zijun Liu
- Green
Preparation Technology of Biobased Materials National & Local
joint Engineering Research Center, Yunnan
Minzu University, Kunming 650500, China
| | - Dengbang Jiang
- Green
Preparation Technology of Biobased Materials National & Local
joint Engineering Research Center, Yunnan
Minzu University, Kunming 650500, China
| | - Mingwei Yuan
- Green
Preparation Technology of Biobased Materials National & Local
joint Engineering Research Center, Yunnan
Minzu University, Kunming 650500, China
| |
Collapse
|
31
|
Scariot DB, Staneviciute A, Machado RRB, Yuk SA, Liu YG, Sharma S, Almunif S, Arona Mbaye EH, Nakamura CV, Engman DM, Scott EA. Efficacy of benznidazole delivery during Chagas disease nanotherapy is dependent on the nanocarrier morphology. Biomaterials 2025; 322:123358. [PMID: 40318604 DOI: 10.1016/j.biomaterials.2025.123358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025]
Abstract
The causative agent of Chagas disease, the protozoan Trypanosoma cruzi, is an obligate intracellular parasite that is typically treated with daily oral administration of Benznidazole (BNZ), a parasiticidal pro-drug with considerable side effects. Previously, we effectively targeted intracellular parasites using ∼100 nm diameter BNZ-loaded poly(ethylene glycol)-b-poly(propylene sulfide) (PEG-b-PPS) vesicular nanocarriers (a.k.a. polymersomes) in a T. cruzi-infected mouse model, without causing the typical side effects associated with standard BNZ treatment. Here, we exploit the structural versatility of the PEG-b-PPS system to investigate the impact of nanocarrier structure on the efficacy of BNZ nanotherapy. Despite sharing the same surface chemistry and oxidation-sensitive biodegradation, solid core ∼25 nm PEG-b-PPS micelles failed to produce in vivo trypanocidal effects. By applying the Förster Resonance Energy Transfer strategy, we demonstrated that PEG-b-PPS polymersomes promoted sustained intracellular drug release and enhanced tissue accumulation, offering a significant advantage for intracellular drug delivery compared to micelles with the same surface chemistry. Our studies further revealed that the lack of parasiticidal effect in PEG-b-PPS micelles is likely due to their slower rate of accumulation into solid tissues, consistent with the prolonged circulation time of intact micelles. Considering the cardiac damage typically induced by T. cruzi infection, this study also investigated the contributions of cardiac cellular biodistribution and payload release for both nanocarriers to the treatment outcomes of BNZ delivery. Our findings emphasize the crucial role of cardiac macrophages in the parasiticidal effect of BNZ formulations and highlight the critical importance of nanobiomaterial structure during therapeutic delivery.
Collapse
Affiliation(s)
- Debora B Scariot
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Austeja Staneviciute
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Rayanne R B Machado
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Biological Sciences, State University of Maringa, Parana, 87020-900, Brazil
| | - Simseok A Yuk
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Yu-Gang Liu
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Swagat Sharma
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Sultan Almunif
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Bioengineering Institute, King Abdulaziz City for Science and Technology, Riyadh, 12354, Saudi Arabia
| | - El Hadji Arona Mbaye
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA
| | - Celso Vataru Nakamura
- Department of Biological Sciences, State University of Maringa, Parana, 87020-900, Brazil
| | - David M Engman
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Pathology, Northwestern University, Chicago, IL, 60611, USA
| | - Evan A Scott
- Department of Biomedical Engineering, Chemistry of Life Processes Institute, Northwestern University, Evanston, IL, 60208, USA; Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA.
| |
Collapse
|
32
|
Ren J, Yan G, Yang L, Kong L, Guan Y, Sun H, Liu C, Liu L, Han Y, Wang X. Cancer chemoprevention: signaling pathways and strategic approaches. Signal Transduct Target Ther 2025; 10:113. [PMID: 40246868 PMCID: PMC12006474 DOI: 10.1038/s41392-025-02167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025] Open
Abstract
Although cancer chemopreventive agents have been confirmed to effectively protect high-risk populations from cancer invasion or recurrence, only over ten drugs have been approved by the U.S. Food and Drug Administration. Therefore, screening potent cancer chemopreventive agents is crucial to reduce the constantly increasing incidence and mortality rate of cancer. Considering the lengthy prevention process, an ideal chemopreventive agent should be nontoxic, inexpensive, and oral. Natural compounds have become a natural treasure reservoir for cancer chemoprevention because of their superior ease of availability, cost-effectiveness, and safety. The benefits of natural compounds as chemopreventive agents in cancer prevention have been confirmed in various studies. In light of this, the present review is intended to fully delineate the entire scope of cancer chemoprevention, and primarily focuses on various aspects of cancer chemoprevention based on natural compounds, specifically focusing on the mechanism of action of natural compounds in cancer prevention, and discussing in detail how they exert cancer prevention effects by affecting classical signaling pathways, immune checkpoints, and gut microbiome. We also introduce novel cancer chemoprevention strategies and summarize the role of natural compounds in improving chemotherapy regimens. Furthermore, we describe strategies for discovering anticancer compounds with low abundance and high activity, revealing the broad prospects of natural compounds in drug discovery for cancer chemoprevention. Moreover, we associate cancer chemoprevention with precision medicine, and discuss the challenges encountered in cancer chemoprevention. Finally, we emphasize the transformative potential of natural compounds in advancing the field of cancer chemoprevention and their ability to introduce more effective and less toxic preventive options for oncology.
Collapse
Affiliation(s)
- Junling Ren
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Guangli Yan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China
| | - Ling Kong
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Yu Guan
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Hui Sun
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
| | - Chang Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Lei Liu
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Ying Han
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China
| | - Xijun Wang
- State key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin, 150040, China.
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou, China.
| |
Collapse
|
33
|
Zhang X, Zhao B, Fu S, Liu Y, Petrisor AA, Yang Z, Fanos HE, Seruya RS, Zhang F. Redox-Responsive Cross-Linking of Polycarbonate Nanomedicines for Enhanced Stability and Controlled Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23135-23145. [PMID: 40173362 DOI: 10.1021/acsami.4c22002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Self-assembled polymeric micelles formed from amphiphilic block copolymers offer a promising strategy for enhanced drug delivery due to their biocompatibility and controlled release. However, challenges such as their poor colloidal stability under diluted conditions and degradation during storage and circulation limit their further applications. To address these issues, we developed a straightforward method for constructing cross-linked polycarbonate micelles that enhance stability while allowing for controlled stimuli-responsive drug delivery. By utilizing disulfide-based cross-linking and covalent conjugation of the anticancer drug, our approach maintains micelle integrity and extremely high drug loading over extended periods as well as the superior control of triggered drug release compared to non-cross-linked versions, demonstrating enhanced stability in complex biological environments and improved anticancer efficacy, presenting a novel platform for stable polymer-drug conjugate nanocarriers, holding significant therapeutic potential for targeted cancer treatment.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Bowen Zhao
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Shiwei Fu
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Yilin Liu
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Ashley A Petrisor
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Zixin Yang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Hannah E Fanos
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Ronald S Seruya
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| | - Fuwu Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
- The Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, 1951 NW seventh Ave, Miami, Florida 33136, United States
| |
Collapse
|
34
|
Wang C, Wu T, Li Y, Liu J, Wang Y, Wang K, Li Y, Leng X. Study of Polyethylene Oxide- b-Poly( ε-caprolactone- ran- δ-valerolactone) Amphiphilic Architectures and Their Effects on Self-Assembly as a Drug Carrier. Polymers (Basel) 2025; 17:1030. [PMID: 40284295 PMCID: PMC12030258 DOI: 10.3390/polym17081030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Amphiphilic block copolymers with complex topologies (e.g., star and brush topologies) have attracted significant attention in drug delivery owing to their superior performance over linear micelles. However, their precise synthesis and structure-property relationships require further investigation. In this study, hydroxylated polybutadiene with adjustable topology and hydroxyl group density was employed as a macroinitiator to synthesize well-defined amphiphilic poly (ethylene oxide)-b-poly(ε-caprolactone-ran-δ-valerolactone) (PEO-b-P(CL-ran-VL)) copolymers via ring-opening polymerization (ROP). A series of linear, star, linear-comb, and star-comb copolymers were prepared as curcumin-loaded micellar carriers for the study. The self-assembly behavior, drug encapsulation efficiency, and in vitro release profiles of these copolymers in aqueous environments were systematically investigated. The results demonstrated that increasing the branch length of star-comb copolymers effectively reduced micelle size from 143 to 96 nm and enhanced drug encapsulation efficiency from 27.3% to 39.8%. Notably, the star-comb architecture exhibited 1.2-fold higher curcumin encapsulation efficiency than the linear counterparts. Furthermore, the optimized star-comb nanoparticles displayed sustained release kinetics (73.38% release over 15 days), outperforming conventional linear micelles. This study establishes a quantitative structure-property relationship between copolymer topology and drug delivery performance, providing a molecular design platform for programmable nanocarriers tailored to diverse therapeutic requirements of various diseases.
Collapse
Affiliation(s)
- Chaoqun Wang
- State Key Laboratory of Fine Chemicals, Liaoning Key Laboratory of Polymer Science and Engineering, Department of Polymer Science and Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (C.W.); (Y.L.); (J.L.); (Y.W.); (K.W.)
| | - Tong Wu
- SINOPEC Ningbo New Materials Research Institute Company Limited, Ningbo 315000, China;
| | - Yidi Li
- State Key Laboratory of Fine Chemicals, Liaoning Key Laboratory of Polymer Science and Engineering, Department of Polymer Science and Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (C.W.); (Y.L.); (J.L.); (Y.W.); (K.W.)
| | - Jie Liu
- State Key Laboratory of Fine Chemicals, Liaoning Key Laboratory of Polymer Science and Engineering, Department of Polymer Science and Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (C.W.); (Y.L.); (J.L.); (Y.W.); (K.W.)
| | - Yanshai Wang
- State Key Laboratory of Fine Chemicals, Liaoning Key Laboratory of Polymer Science and Engineering, Department of Polymer Science and Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (C.W.); (Y.L.); (J.L.); (Y.W.); (K.W.)
| | - Kefeng Wang
- State Key Laboratory of Fine Chemicals, Liaoning Key Laboratory of Polymer Science and Engineering, Department of Polymer Science and Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (C.W.); (Y.L.); (J.L.); (Y.W.); (K.W.)
| | - Yang Li
- State Key Laboratory of Fine Chemicals, Liaoning Key Laboratory of Polymer Science and Engineering, Department of Polymer Science and Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (C.W.); (Y.L.); (J.L.); (Y.W.); (K.W.)
| | - Xuefei Leng
- State Key Laboratory of Fine Chemicals, Liaoning Key Laboratory of Polymer Science and Engineering, Department of Polymer Science and Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (C.W.); (Y.L.); (J.L.); (Y.W.); (K.W.)
| |
Collapse
|
35
|
Rasoulianboroujeni M, Kang RH, Klukas M, Kwon GS. Crystallization of supersaturated PEG-b-PLA for the production of drug-loaded polymeric micelles. J Control Release 2025; 380:457-468. [PMID: 39921034 PMCID: PMC11908913 DOI: 10.1016/j.jconrel.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/19/2024] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
In this study, we propose the "crystallization from supersaturated solution" method for producing drug-loaded polymeric micelles. This method involves the formation of solid drug-encapsulating crystals of a diblock copolymer through isothermal crystallization from a supersaturated solution of the copolymer in low molecular weight PEGs containing the drug, followed by dissolution of the crystals to obtain drug-loaded micelles. We fabricated and characterized micelles loaded with several model drugs (paclitaxel, rapamycin, and docetaxel) and their oligo(lactic acid)8-prodrugs using PEG4kDa-b-PLA2.2kDa as the micelle-forming copolymer and PEGs of varying molecular weights (200, 400, and 600 Da) as solvents. Our findings indicate that the molecular weight of the solvent PEG and the target drug loading significantly influence the physicochemical properties of the resulting micelles, including loading efficiency and particle size distribution. Micelles produced with PEG200 as the solvent exhibited the highest loading efficiency, followed by those made with PEG600 and PEG400 for all the drugs and prodrugs tested. Increasing the target drug loading enhanced both the loading efficiency and average particle size across all formulations. Furthermore, prodrug-loaded micelles showed higher loading efficiency and improved stability in aqueous solutions compared to their parent drug counterparts. Crystals encapsulating both parent drugs and prodrugs could be stored at room temperature for extended periods, producing micelles with no significant differences in loading efficiency and particle size distribution compared to freshly prepared micelles. Additionally, the crystals demonstrated a rapid dissolution rate, forming uniform micelles after just 5 s of hydration and agitation. Cytotoxicity studies against 4 T1 and MDA-MB-231 breast cancer cell lines revealed that the molecular weight of the PEG used as the solvent impacts the cytotoxicity of the resulting micelles, with those produced using PEG200 displaying the highest cytotoxicity, followed by PEG400 and PEG600. Overall, the crystallization from supersaturated solution method proves to be an effective platform for prolonged storage and rapid formation of stable, drug-loaded polymeric micelles. It has the potential to eliminate the need for freeze-drying in the formulation and storage of drug-loaded polymeric micelles. These findings highlight the method's potential for advancing drug delivery systems, particularly for the solubilization of hydrophobic drugs using micellar formulations.
Collapse
Affiliation(s)
- Morteza Rasoulianboroujeni
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States
| | - Rae Hyung Kang
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States; Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Maraya Klukas
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States
| | - Glen S Kwon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI 53705, United States.
| |
Collapse
|
36
|
Ricci-Junior E, Rosa AS, do Nascimento T, Santos-Oliveira R, da Silva MAN, Barreto-Vieira DF, Batista LT, da Conceição GB, Quintão TAN, Ferreira VNS, Miranda MD. Nanotechnology-Driven Strategy Against SARS-CoV-2: Pluronic F127-Based Nanomicelles with or Without Atazanavir Reduce Viral Replication in Calu-3 Cells. Viruses 2025; 17:518. [PMID: 40284961 PMCID: PMC12031194 DOI: 10.3390/v17040518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Despite extensive efforts, no highly effective antiviral molecule exists for treating moderate and severe COVID-19. Nanotechnology has emerged as a promising approach for developing novel drug delivery systems to enhance antiviral efficacy. Among these, polymeric nanomicelles improve the solubility, bioavailability, and cellular uptake of therapeutic agents. In this study, Pluronic F127-based nanomicelles were developed and evaluated for their antiviral activity against SARS-CoV-2. The nanomicelles, formulated using the direct dissolution method, exhibited an average size of 37.4 ± 8.01 nm and a polydispersity index (PDI) of 0.427 ± 0.01. Their antiviral efficacy was assessed in SARS-CoV-2-infected Vero E6 and Calu-3 cell models, where treatment with a 1:2 dilution inhibited viral replication by more than 90%. Cytotoxicity assays confirmed the nanomicelles were non-toxic to both cell lines after 72 h. In SARS-CoV-2-infected Calu-3 cells (human type II pneumocyte model), treatment with Pluronic F127-based nanomicelles containing atazanavir (ATV) significantly reduced viral replication, even under high MOI (2) and after 48 h, while also preventing IL-6 upregulation. To investigate their mechanism, viral pretreatment with nanomicelles showed no inhibitory effect. However, pre-exposure of Calu-3 cells led to significant viral replication reduction (>85% and >75% for 1:2 and 1:4 dilutions, respectively), as confirmed by transmission electron microscopy. These findings highlight Pluronic F127-based nanomicelles as a promising nanotechnology-driven strategy against SARS-CoV-2, reinforcing their potential for future antiviral therapies.
Collapse
Affiliation(s)
- Eduardo Ricci-Junior
- Galenic Development Laboratory, University Pharmacy, Universidade Federal de Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Alice Santos Rosa
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Tatielle do Nascimento
- Galenic Development Laboratory, University Pharmacy, Universidade Federal de Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmacy and Synthesis of Novel Radiopharmaceuticals, Nuclear Engineering Institute, Rio de Janeiro 21941-906, Brazil;
- Laboratory of Nanoradiopharmacy and Radiopharmaceuticals, Zona Oeste State University, Rio de Janeiro 21941-906, Brazil
| | - Marcos Alexandre Nunes da Silva
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Medicina Tropical, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Debora Ferreira Barreto-Vieira
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
- Programa de Pós-graduação em Medicina Tropical, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| | - Luísa Tozatto Batista
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Giovanna Barbosa da Conceição
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Tayane Alvites Nunes Quintão
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Vivian Neuza Santos Ferreira
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
| | - Milene Dias Miranda
- Laboratory of Morphology and Viral Morphogenesis, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, Brazil; (A.S.R.); (M.A.N.d.S.); (D.F.B.-V.); (L.T.B.); (G.B.d.C.); (T.A.N.Q.); (V.N.S.F.)
- Programa de Pós-graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
37
|
Santander Plantamura YA, Allo M, Riedel J, Fuentes P, Riesco AS, Bernabeu E, Garcés M, Evelson P, Gorzalczany S, Carranza A, Höcht C, Chiappetta D. Development of a new micellar formulation of carvedilol and curcumin to enhance blood pressure reduction in a spontaneously hypertensive rat model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4165-4177. [PMID: 39422748 DOI: 10.1007/s00210-024-03537-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Cardiovascular diseases remain a leading cause of morbidity and mortality worldwide, requiring innovative therapeutic strategies. This project explores a nano-pharmaceutical approach to enhance the efficacy of cardiovascular drugs, focusing on carvedilol and curcumin. These agents, known for their potential cardiovascular benefits, are encapsulated within Soluplus® micelles to form a novel drug delivery system. The novelty of this formulation lies in its ability to significantly improve the solubility of both carvedilol and curcumin, which have traditionally been limited by their hydrophobic nature. By utilizing Soluplus® micelles, we have developed a unique delivery system that optimizes the therapeutic potential of both drugs. The nanomicelles were meticulously characterized for drug loading, size distribution, and morphological features. The carvedilol and curcumin release patterns were investigated, revealing sustained and controlled release profiles. Additionally, the antioxidant capacity of the micellar formulation was evaluated, demonstrating the preservation of curcumin's antioxidative properties. In vivo studies using spontaneously hypertensive male rats explored the pharmacokinetics and hemodynamic effects of the nanomicellar system. These results indicated successful encapsulation of both drugs without altering their plasma profiles. Furthermore, the administration of carvedilol and curcumin micelles exhibited a more significant reduction in mean arterial pressure compared to individual drug administration, suggesting a potential synergistic effect. In conclusion, this nano-pharmaceutical approach offers a promising avenue for cardiovascular therapy, providing a platform for combined drug delivery and potential synergistic effects. The optimized formulation could lead to improved patient outcomes and enhanced cardiovascular health.
Collapse
Affiliation(s)
- Yanina Alejandra Santander Plantamura
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina.
- Unidad de Investigación en Biomedicina, Facultad de Estudios Superiores Iztacala (FES-Iztacala), Universidad Nacional Autónoma de México (UNAM), Tlalnepantla, Edo. de México, México.
| | - Miguel Allo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina
| | - Jennifer Riedel
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Tecnología Farmacéutica I, Buenos Aires, Argentina
| | - Pedro Fuentes
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Tecnología Farmacéutica I, Buenos Aires, Argentina
| | - Ana Sol Riesco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina
| | - Ezequiel Bernabeu
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Tecnología Farmacéutica I, Buenos Aires, Argentina
- Universidad de Buenos Aires, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentina
| | - Mariana Garcés
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Química General E Inorgánica, Buenos Aires, Argentina
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Química General E Inorgánica, Buenos Aires, Argentina
- Universidad de Buenos Aires, CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Susana Gorzalczany
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina
| | - Andrea Carranza
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Christian Höcht
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Farmacología, Buenos Aires, Argentina
- Universidad de Buenos Aires, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentina
| | - Diego Chiappetta
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Tecnología Farmacéutica I, Buenos Aires, Argentina
- Universidad de Buenos Aires, Instituto de Tecnología Farmacéutica y Biofarmacia (InTecFyB), Buenos Aires, Argentina
| |
Collapse
|
38
|
Jiang X, Gong M, Jia Y, Adu-Frimpong M, Wang X, Hua Q, Li T, Li J, Pan P, Toreniyazov E, Yu J, Cao X, Wang Q, Xu X. Preparation, in vitro and in vivo evaluation and anti-renal injury effects of Niazimicin-loaded mixed polymeric micelles. J Pharm Sci 2025; 114:103703. [PMID: 39988296 DOI: 10.1016/j.xphs.2025.103703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Chronic Kidney Disease (CKD) has become one of the major life-threatening conditions. Moringa seeds have been reported to exhibit renoprotective effects, with Niazimicin as its characteristic component. OBJECTIVE To investigate the anti-renal injury effects of Niazimicin and its mixed micelles (N-M) that composed of monomethyl ether poly (ethylene glycol)-polycaprolactone (mPEG-PCL) and polyethylene glycolated chitosan (PEG-CS) on adenine-induced CKD mice. METHODS PEG-CS was prepared via formaldehyde linkage method. The thin film dispersion method was employed for the preparation of N-M before it was characterized in vivo and in vitro. The anti-renal injury effects were evaluated by analyzing the serum levels of creatinine (Cr), p-Cresol sulphate (pCs), indole sulphate (IS) and hematoxylin-eosin (HE)-stained sections of hepatic and renal pathological tissues in CKD mice. RESULTS The N-M were spherical micelles of uniform size and highly dispersed with particle size of 42.94 ± 0.58 nm, encapsulation efficiency (EE) of 97.73 ± 2.33% and drug loading (DL) of 16.17 ± 0.28%, as well as good stability, and a very low critical micelle concentration (CMC) value of 0.00731 mg/mL. The N-M had a delayed-release effect and higher oral bioavailability compared to Niazimicin. CONCLUSION In CKD mice, Niazimicin exhibited an anti-renal injury effect, while the renoprotective effect of N-M was superior to that of Niazimicin.
Collapse
Affiliation(s)
- Xia Jiang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Mingie Gong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Yue Jia
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School Chemical and Biochemical Sciences, C.K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, UK, 0215-5321, Ghana
| | - Xiaowen Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Qinyang Hua
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Tingyuan Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Jiaying Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Pengfei Pan
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | | | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| |
Collapse
|
39
|
Wang F, Zeng J, Lin L, Wang X, Zhang L, Tao N. Co-delivery of astaxanthin using positive synergistic effect from biomaterials: From structural design to functional regulation. Food Chem 2025; 470:142731. [PMID: 39755039 DOI: 10.1016/j.foodchem.2024.142731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/22/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
The powerful antioxidant properties of astaxanthin (AST) face two significant challenges: low water solubility and poor chemical stability. To overcome them, extensive research and development efforts have been directed toward creating effective delivery systems. Among them, the positive synergistic effect between biomaterials can be used to refine the design of delivery systems. Understanding the relationship between structure and function aids in tailoring applications to specific needs. This review outlines the challenges associated with delivering AST and reviews the mechanisms involved in creating delivery systems, specifically focusing on the structure-function relationship of biomaterials. It comprehensively introduces the positive synergistic effect of biomaterials with enhancing the functional properties of AST, and analyzes the impact of designed structures on function regulation and the application prospects of the delivery system in the food industry. The future demand for efficient delivery of AST will increasingly depend on the positive synergistic effect between biomaterials.
Collapse
Affiliation(s)
- Fengqiujie Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Jianhua Zeng
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Liu Lin
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Xichang Wang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquatic-Product Processing and Preservation, Shanghai, China; National Experimental Teaching Demonstration Center for Food Science and Engineering, Shanghai Ocean University, Shanghai, China
| | - Long Zhang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquatic-Product Processing and Preservation, Shanghai, China; National Experimental Teaching Demonstration Center for Food Science and Engineering, Shanghai Ocean University, Shanghai, China.
| | - Ningping Tao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquatic-Product Processing and Preservation, Shanghai, China; National Experimental Teaching Demonstration Center for Food Science and Engineering, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
40
|
Caporusso A, Radice M, Biundo A, Gorgoglione R, Agrimi G, Pisano I. Waste cooking oils as a sustainable feedstock for bio-based application: A systematic review. J Biotechnol 2025; 400:48-65. [PMID: 39952410 DOI: 10.1016/j.jbiotec.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Waste cooking oils (WCOs) are common wastes and promising green, eco-friendly and sustainable feedstocks for bio-based applications. While the primary valorisation strategy revolves around the concept of waste-to energy, new research trends have emerged in the last decade. This systematic review provides a comprehensive analysis of the current state of the art in the conversion of WCOs into bio-based molecules. Based on the PRISMA methodology, 64 papers were selected using different databases and sources, such as: PubMed, ScienceDirect, Scopus and MDPI. The data extraction process focused on studies reporting the biological and chemical conversion of WCOs into value-added bioproducts. Many of the selected publications deal with the development of bioactive molecules, including biosurfactants, with application in pharmaceuticals, food, cosmetics, and bioremediation. Bioconversion processes mainly featured engineered Yarrowia lipolytica and Escherichia coli strains, even if additional microorganisms were also employed. In the same way, different chemical processes have been thoroughly studied. A smaller segment of research is directed to the production of feed supplements and soaps. Regulatory constraints limit further development in feed supplements due to potential contaminants, while soap production needs further stability studies. The present systematic review shows promising outcomes in the valorisation of WCOs through the development of value-added molecules and products. Despite the wide range of applications, these findings identify that the scalability and economic sustainability of the selected processes require further investigation. This study seeks to summarize the current state of the art and identify potential gaps to advance the industrialization of WCOs valorisation.
Collapse
Affiliation(s)
- Antonio Caporusso
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via E. Orabona, 4, Bari 70125, Italy
| | - Matteo Radice
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via E. Orabona, 4, Bari 70125, Italy.
| | - Antonino Biundo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via E. Orabona, 4, Bari 70125, Italy; REWOW srl, Via G. Matarrese 10, Bari 70124, Italy
| | - Ruggiero Gorgoglione
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via E. Orabona, 4, Bari 70125, Italy
| | - Gennaro Agrimi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via E. Orabona, 4, Bari 70125, Italy; CIRCC, Interuniversity Consortium Chemical Reactivity and Catalysis, Via C. Ulpiani, 27, Bari 70126, Italy
| | - Isabella Pisano
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, Via E. Orabona, 4, Bari 70125, Italy; CIRCC, Interuniversity Consortium Chemical Reactivity and Catalysis, Via C. Ulpiani, 27, Bari 70126, Italy.
| |
Collapse
|
41
|
Li W, Antoniadi L, Zhou H, Chen H, Angelis A, Halabalaki M, Skaltsounis LA, Qi Z, Wang C. Sodium cholate-coated Olea europaea polyphenol nanoliposomes: Preparation, stability, release, and bioactivity. Food Chem 2025; 469:142580. [PMID: 39721438 DOI: 10.1016/j.foodchem.2024.142580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Ultra-flexible nanoliposomes (UNL) coated with sodium cholate were fabricated using the thin film hydration technique to encapsulate oleocanthal (OLEO), oleacein (OLEA), oleuropein (OLEU), and hydroxytyrosol (HT) for improving their stability and bioactivity. Their physicochemical properties were further validated through DLS, FTIR, XRD, TGA, and DSC analyses. Negative-staining TEM imaging revealed well-dispersed UNL with laminar vesicles inside. Additionally, their transdermal studies in vitro demonstrated that UNL enhanced the cumulative release of OLEO, OLEA, OLEU, and HT by 3.13, 2.76, 2.59, and 2.83 times, respectively. Furthermore, their release mechanisms were better approximated the Peppas-Sahlin model rather than the Korsmeyer-Peppas and Higuchi models, which governed by Fickian diffusion. Moreover, comparing to their compounds, UNL structure exhibited improved their antioxidant and cytotoxicity properties, highlighting their potential as effective delivery agents in humans. These results offer a novel approach for stabilizing biologically active polyphenols from Olea europaea, paving the way for enhanced human health applications.
Collapse
Affiliation(s)
- Wenjun Li
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Laboratory for Biomass Chemical Utilization, Key and Open Laboratory on Forest Chemical Engineering, SFA, Key Laboratory of Biomass Energy and Material, Nanjing 210042, Jiangsu Province, China
| | - Lemonia Antoniadi
- Division of Pharmacognosy and Natural Products Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli Zografou, NKUA, 15771, Athens, Greece; Pharmagnose S.A., 57th km Athens-lamia National Road, Oinofyta 32011, Greece
| | - Hao Zhou
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Laboratory for Biomass Chemical Utilization, Key and Open Laboratory on Forest Chemical Engineering, SFA, Key Laboratory of Biomass Energy and Material, Nanjing 210042, Jiangsu Province, China
| | - Hongxia Chen
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Laboratory for Biomass Chemical Utilization, Key and Open Laboratory on Forest Chemical Engineering, SFA, Key Laboratory of Biomass Energy and Material, Nanjing 210042, Jiangsu Province, China
| | - Apostolis Angelis
- Division of Pharmacognosy and Natural Products Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli Zografou, NKUA, 15771, Athens, Greece
| | - Maria Halabalaki
- Division of Pharmacognosy and Natural Products Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli Zografou, NKUA, 15771, Athens, Greece
| | - Leandros A Skaltsounis
- Division of Pharmacognosy and Natural Products Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli Zografou, NKUA, 15771, Athens, Greece
| | - Zhiwen Qi
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Laboratory for Biomass Chemical Utilization, Key and Open Laboratory on Forest Chemical Engineering, SFA, Key Laboratory of Biomass Energy and Material, Nanjing 210042, Jiangsu Province, China.
| | - Chengzhang Wang
- Institute of Chemical Industry of Forest Products, CAF, National Engineering Laboratory for Biomass Chemical Utilization, Key and Open Laboratory on Forest Chemical Engineering, SFA, Key Laboratory of Biomass Energy and Material, Nanjing 210042, Jiangsu Province, China.
| |
Collapse
|
42
|
Floyd TG, Gurnani P, Rho JY. Characterisation of polymeric nanoparticles for drug delivery. NANOSCALE 2025; 17:7738-7752. [PMID: 40018862 DOI: 10.1039/d5nr00071h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Polymeric nanoparticles represent an innovative approach to drug delivery, particularly for addressing complex diseases like cancer. Their nanoscale dimensions facilitate targeted cellular uptake and effective navigation of biological barriers. With a broad range of polymerisation and functionalisation techniques, these nanoparticles can enable precise drug release, enhanced stability, and improved bioavailability while minimising side effects. Compared to conventional carriers, polymeric nanoparticles offer superior stability and versatility. However, despite these beneficial attributes, challenges remain in understanding their dynamic behaviour and interactions within biological systems. This mini-review aims to highlight key characterisation methods for studying polymeric nanocarriers, explore recent advances, and examine current challenges that must be addressed to optimise their therapeutic potential and advance these promising targeted drug delivery systems.
Collapse
Affiliation(s)
- Thomas G Floyd
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Pratik Gurnani
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, Bloomsbury, London, WC1N 1AX, UK
| | - Julia Y Rho
- Department of Chemistry, University College London, 20 Gordon Street, London, WC1H 0AJ, UK.
| |
Collapse
|
43
|
De Caro L, Stoll T, Grandeury A, Gozzo F, Giannini C. Characterization of VitE-TPGS Micelles Linked to Poorly Soluble Pharmaceutical Compounds Exploiting Pair Distribution Function's Moments. Pharmaceutics 2025; 17:431. [PMID: 40284426 PMCID: PMC12030118 DOI: 10.3390/pharmaceutics17040431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Micelles have attracted significant interest in nanomedicine as drug delivery systems. This study investigates the morphology of micelles formed by the D-α-tocopherol polyethylene glycol 1000 succinate (VitE-TPGS) surfactant in the presence and absence of, respectively, a poorly soluble pharmaceutical compound (PSC), i.e., Eltrombopag (0.08 wt%) and CaCl2 (0.03 wt%). The aim was to assess the micelles' ability to solubilize the PSC and potentially shield it from Ca2+ ions, simulating in vivo conditions. Methods: For this purpose, we have developed a novel theoretical approach for analyzing Pair Distribution Function (PDF) data derived from Small-Angle X-ray Scattering (SAXS) measurements, based on the use of PDF's moments. Results: Our spheroid-based model was able to characterize successfully the micellar morphology and their interactions with PSC and CaCl2, providing detailed insights into their size, shape, and electron density contrasts. The presence of PSC significantly affected the shape and integral of the PDF curves, indicating incorporation into the micelles. This also resulted in a decrease in the micelle size, regardless of the presence of CaCl2. When this salt was added, it reduced the amount of PSC within the micelles. This is likely due to a decrease in the overall PSC availability in solution, induced by Ca2+ ions. Conclusions: This advanced yet straightforward analytical model represents a powerful tool for characterizing and optimizing micelle-based drug delivery systems.
Collapse
Affiliation(s)
- Liberato De Caro
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Via Amendola 122/O, 70125 Bari, Italy;
| | - Thibaud Stoll
- Excelsus Structural Solutions (Swiss) AG, Park Innovaare, Parkstrasse 1, 5234 Villigen, Switzerland; (T.S.); (F.G.)
| | - Arnaud Grandeury
- Novartis Pharma AG, Technical Research and Development, Material Science, Novartis Campus, Virchow 6.3.231, 4056 Basel, Switzerland;
| | - Fabia Gozzo
- Excelsus Structural Solutions (Swiss) AG, Park Innovaare, Parkstrasse 1, 5234 Villigen, Switzerland; (T.S.); (F.G.)
| | - Cinzia Giannini
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Via Amendola 122/O, 70125 Bari, Italy;
| |
Collapse
|
44
|
Shah T, Polara H, Babanyinah G, Bhadran A, Wang H, Castillo CC, Grabowski G, Biewer MC, Torabifard H, Stefan MC. Computational design to experimental validation: molecular dynamics-assisted development of polycaprolactone micelles for drug delivery. J Mater Chem B 2025; 13:4166-4178. [PMID: 40047718 DOI: 10.1039/d4tb02789b] [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: 03/27/2025]
Abstract
Amphiphilic diblock copolymers are used in drug delivery systems for cancer treatments. However, these carriers suffer from lower drug loading capacity, poor water solubility, and non-targeted drug release. Here, we utilized a computational approach to analyze the effect of the functional groups of the hydrophobic block on the drug-polymer interactions. To design effective drug carriers, four different amphiphilic block copolymer micelles with distinct aromatic and heteroaromatic groups at the hydrophobic core were subjected to molecular dynamics simulations. The solvent-accessible surface area, water shell, hydrogen bonding, and radius of gyration of the simulated micelles were determined. Further, we assessed the interactions between the hydrophobic block and drug molecules using linear interaction energy and non-covalent interactions. The computational studies revealed that the micelles containing a novel poly(γ-2-methoxyfuran-ε-caprolactone) (PFuCL) hydrophobic block have the highest polymer-drug interactions. From these findings, we synthesized a novel amphiphilic poly(ethylene glycol)-b-poly(γ-2-methoxyfuran(ε-caprolactone)) (PEG-b-PFuCL) block copolymer using ring-opening polymerization of FuCL monomer. The polymer was self-assembled in aqueous media to form micelles. The aromatic segment of PEG-b-PFuCL micelles enhanced the doxorubicin (DOX) loading through non-covalent interactions, resulting in a 4.25 wt% drug-loading capacity. We also showed that the hydrolysis of the ester bond allowed a faster in vitro drug release at pH 5.0 compared to pH 7.4. Cell viability experiments revealed that DOX-loaded PEG-b-PFuCL micelles show that micelles are cytotoxic and readily uptaken into MDA-MB-231 cells. Therefore, furan-substituted micelles will be an ideal drug carrier with higher polymer-to-drug interactions, enhanced drug loading, and lower premature leakage.
Collapse
Affiliation(s)
- Tejas Shah
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Himanshu Polara
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Godwin Babanyinah
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Abhi Bhadran
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Hanghang Wang
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Cristina Cu Castillo
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Gerik Grabowski
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Michael C Biewer
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Hedieh Torabifard
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| | - Mihaela C Stefan
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA.
| |
Collapse
|
45
|
Shaikh S, Chary PS, Mehra NK. Tyrosine Kinase Inhibitor Lenvatinib Based Nano Formulations and Cutting-Edge Scale-Up Technologies in revolutionizing Cancer Therapy. ACS APPLIED BIO MATERIALS 2025; 8:1749-1784. [PMID: 40091597 DOI: 10.1021/acsabm.4c01527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Lenvatinib (LEN), a tyrosine kinase inhibitor, has emerged as a promising therapeutic agent for various solid tumors. Nevertheless, a number of constraints, including diminished bioavailability, incapacity to elicit localized inflammation, and inability to selectively accumulate at the tumor site, may impede the comprehensive exploitation of its versatile tyrosine kinase inhibitory capabilities. In order to achieve targeted delivery of LEN while also reducing its high dose used in conventional therapeutics, nanoformulation approaches can be adopted. The integration of LEN into various nanoformulations, such as nanoparticles, nanocrystals, high density lipoproteins (HDLs), liposomes, and micelles, is discussed, highlighting the advantages of these innovative approaches in a comparative manner; however, given that the current methods of nanoformulation synthesis employ toxic organic solvents and chemicals, there is an imperative need for exploring alternative, environmentally friendly approaches. The multifaceted effects of nanocarriers have rendered them profoundly applicable within the biomedical domain, serving as instrumental entities in various capacities such as vehicles for drug delivery and genetic material, diagnostic agents, facilitators of photothermal therapy, and radiotherapy. However, the scalability of these nanotechnological methodologies must be rigorously investigated and addressed to refine drug delivery mechanisms. This endeavor offers promising prospects for revolutionizing strategies in cancer therapeutics, thereby laying the foundation for future research in scale-up techniques in the pursuit of more effective and less toxic therapies for cancer.
Collapse
Affiliation(s)
- Samia Shaikh
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Padakanti Sandeep Chary
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500029, India
| |
Collapse
|
46
|
Zurletti B, Andreana I, Salaroglio IC, Bincoletto V, Manzoli M, Rolando B, Milla P, Riganti C, Stella B, Arpicco S. Tailoring the Composition of HA/PEG Mixed Nano-Assemblies for Anticancer Drug Delivery. Molecules 2025; 30:1349. [PMID: 40142123 PMCID: PMC11945053 DOI: 10.3390/molecules30061349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
Self-assembling amphiphilic polymers represent highly promising materials with emerging applications across various fields. In these polymers, the presence of hydrophilic and hydrophobic segments within their structure drives the self-assembly process in aqueous environments, leading to organized structures capable of incorporating lipophilic drugs. Their high chemical versatility enables the design of tailored structures to meet specific requirements, such as the active targeting ability, thereby broadening their potential applications. In this work, a polyethylene glycol-phospholipid conjugate was employed to form nanocarriers loaded with a lipophilic derivative of gemcitabine. To achieve nano-assemblies actively targeted towards cancer cells overexpressing the hyaluronic acid (HA) receptor CD44, a HA-phospholipid conjugate was co-formulated in various molar ratios (1%, 10%, and 20%). All formulations exhibited a mean diameter below 130 nm, a negative zeta potential (approximately -30 mV), and a high encapsulation efficiency (above 90%). These nano-assemblies demonstrated stability during storage and effectively released the encapsulated drug in a cell culture medium. Upon incubation with cancer cells, the nano-assemblies were internalized via a CD44 endocytosis-mediated mechanism, with the extent of internalization depending on the HA conjugate content. Consistently, cell viability studies revealed that the nanocarriers decorated with higher amounts of HA exerted a higher cytotoxicity, enabling a fine tuning of the nano-assembly properties.
Collapse
Affiliation(s)
- Beatrice Zurletti
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| | - Ilaria Andreana
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| | - Iris Chiara Salaroglio
- Department of Oncology, Interdepartmental Center of Molecular Biotechnology “Guido Tarone”, University of Turin, Via Nizza 44, 10126 Turin, Italy; (I.C.S.); (C.R.)
| | - Valeria Bincoletto
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| | - Maela Manzoli
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| | - Barbara Rolando
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| | - Paola Milla
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| | - Chiara Riganti
- Department of Oncology, Interdepartmental Center of Molecular Biotechnology “Guido Tarone”, University of Turin, Via Nizza 44, 10126 Turin, Italy; (I.C.S.); (C.R.)
| | - Barbara Stella
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy; (B.Z.); (I.A.); (V.B.); (M.M.); (B.R.); (P.M.); (B.S.)
| |
Collapse
|
47
|
Lang W, Watanabe T, Lee C, Tagami T, Li F, Yamamoto T, Tajima K, Borsali R, Takahashi K, Satoh T, Isono T. Fully biosourced amphiphilic block copolymer from tamarind seed xyloglucan and solanesol: synthesis, aqueous self-assembly, and drug encapsulation. Carbohydr Polym 2025; 352:123181. [PMID: 39843085 DOI: 10.1016/j.carbpol.2024.123181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025]
Abstract
This study aims to explore the development of natural bio-based amphiphilic block copolymers for drug delivery applications. We investigated block copolymers derived from tamarind seed xyloglucan and solanesol, focusing on their synthesis, structural analysis, aqueous self-assembly, and drug encapsulation. Specifically, xyloglucan hydrolysate segments with number-average degrees of polymerization (DPs) of between 8 and 44 (XOS8, XMS10, XMS16, XMS28, and XMS44) were used as the hydrophilic blocks, whereas plant-sourced solanesol was selected as the hydrophobic segment. These two segments were linked via click chemistry to create novel XOS-b-Sol and XMS-b-Sol copolymers, where XOS refers to a xyloglucan oligosaccharide (DP < 10), and XMS denotes a megalosaccharide, defined as a larger xyloglucan polymer with a DP >10. XMS10- and XMS16-b-Sol self-assembled in aqueous media to form predominantly narrowly dispersed spherical micelles, while XOS8-b-Sol exhibited a combination of short wormlike structures and spherical micelles. Small-angle X-ray scattering, multi-angle dynamic light scattering, and transmission electron microscopy revealed the XMS28- and XMS44-b-Sol micelles are morphologically diverse. XMS10-b-Sol solubilized quercetin, a water-insoluble flavonoid, highly effectively, highlighting its potential as an environmentally benign drug carrier.
Collapse
Affiliation(s)
- Weeranuch Lang
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Tomohisa Watanabe
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Chaehun Lee
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Takayoshi Tagami
- Research Faculty of Agriculture, Hokkaido University, Sapporo 060-8589, Japan
| | - Feng Li
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Takuya Yamamoto
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Kenji Tajima
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | | | - Kenji Takahashi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Kanazawa 920-1192, Japan
| | - Toshifumi Satoh
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan; ICReDD List-PF, Hokkaido University, N21W10, Kita-ku, Sapporo 001-0021, Japan; Department of Chemical & Materials Engineering, National Central University, Taoyuan 320317, Taiwan.
| | - Takuya Isono
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan.
| |
Collapse
|
48
|
Narayana S, Gowda BHJ, Hani U, Ahmed MG, Asiri ZA, Paul K. Smart Poly(N-isopropylacrylamide)-Based Hydrogels: A Tour D'horizon of Biomedical Applications. Gels 2025; 11:207. [PMID: 40136912 PMCID: PMC11942434 DOI: 10.3390/gels11030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Hydrogels are innovative materials characterized by a water-swollen, crosslinked polymeric network capable of retaining substantial amounts of water while maintaining structural integrity. Their unique ability to swell or contract in response to environmental stimuli makes them integral to biomedical applications, including drug delivery, tissue engineering, and wound healing. Among these, "smart" hydrogels, sensitive to stimuli such as pH, temperature, and light, showcase reversible transitions between liquid and semi-solid states. Thermoresponsive hydrogels, exemplified by poly(N-isopropylacrylamide) (PNIPAM), are particularly notable for their sensitivity to temperature changes, transitioning near their lower critical solution temperature (LCST) of approximately 32 °C in water. Structurally, PNIPAM-based hydrogels (PNIPAM-HYDs) are chemically versatile, allowing for modifications that enhance biocompatibility and functional adaptability. These properties enable their application in diverse therapeutic areas such as cancer therapy, phototherapy, wound healing, and tissue engineering. In this review, the unique properties and behavior of smart PNIPAM are explored, with an emphasis on diverse synthesis methods and a brief note on biocompatibility. Furthermore, the structural and functional modifications of PNIPAM-HYDs are detailed, along with their biomedical applications in cancer therapy, phototherapy, wound healing, tissue engineering, skin conditions, ocular diseases, etc. Various delivery routes and patents highlighting therapeutic advancements are also examined. Finally, the future prospects of PNIPAM-HYDs remain promising, with ongoing research focused on enhancing their stability, responsiveness, and clinical applicability. Their continued development is expected to revolutionize biomedical technologies, paving the way for more efficient and targeted therapeutic solutions.
Collapse
Affiliation(s)
- Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India;
| | - B. H. Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India;
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (U.H.); (Z.A.A.)
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India;
| | - Zahrah Ali Asiri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia; (U.H.); (Z.A.A.)
| | - Karthika Paul
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India;
| |
Collapse
|
49
|
Pandey S, Patel K, Gupta A, Joshi S, Yadav JS, Tripathi P. Pluronics® F68 and D-α-tocopheryl polyethylene glycol succinate 1000 tailored self-assembled mixed micelles to improve oral bioavailability of oleanolic acid: in vitro and in vivo characterization. Drug Deliv Transl Res 2025:10.1007/s13346-025-01834-8. [PMID: 40082366 DOI: 10.1007/s13346-025-01834-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Oleanolic acid (OA) ischaracterized by its low water solubility, poor permeability and majorly metabolized by cytochrome P450 (CYP) isozymes in the intestinal tract, particularly CYP3A, which contribute to the low oral bioavailability. OA has multiple pharmacological actions including hepatoprotective, anti-inflammatory, antidiabetic and antiviral effects. OA classified as a BCS IV drug which have restricted its potential clinical application. In this study D-α-Tocopheryl polyethylene glycol succinate (TPGS) and Pluronics F68 based stabilized OA loaded mixed micellar system (OA-MMs) developed to improve the solubility and permeability. Mixed micelles were characterized by dynamic light scattering studies as a function of temperature, salt addition, and OA solubilisation followed byXRD, FE-SEM and IR analysis confirmed the formation of stabilized OA-MMs with the least size and PDI (10.041 ± 1.35 nm, 0.313 ± 0.012). Scattering studies results demonstrates the formation of stable micelles with no significant alterations insize upon salt addition (up to 150mM NaCl), OA incorporation (up to 150 mM) and temperature rise till 40 °C.Solubility of the pure OA and OA-MMs was found to be 0.042 mg/ml and 1.98 mg/ml. The % cumulative release of drug from alone OA, OA + TPGS and OA-MMs was found to be 4.363 ± 0.025%, 57.18 ± 0.034% and 92.269 ± 0.017% respectively up to 24 h. Single-pass intestinal perfusion studies (SPIP) showed that Ka and Peffective of OA-MMs was improved30 fold as compared with that of pure OA and this was mainly due to the improved permeability and inhibitory effect of Pluronic F68 on CYP3A. The in vivo Pharmacokinetic study showed that Cmax increased markedly from 12.76 to 20.49 and 39.17 µg/ml in case of OA alone, OA + TPGS and OA-MMs. Parallel to the Cmax there was an increase in the AUC0-24133.68 to 164.56 and 296.50 respectively. All of the produced OA-MMs formulation's results demonstrated a notable increase in OA's bioavailability through increased permeability and solubility along with metabolic inhibition OA.
Collapse
Affiliation(s)
- Sonia Pandey
- Department of Pharmacy, Yashraj College of Professional Studies, Kanpur, Uttar Pradesh, 209217, India.
| | - Komal Patel
- Maliba Pharmacy College, Uka Tarsadia University, Surat, 394350, India
| | - Arti Gupta
- Shri Ram Murti Smarak College of Engineering and Technology (Pharmacy), Bareilly, Uttar Pradesh, 243202, India
| | - Shrikant Joshi
- Maliba Pharmacy College, Uka Tarsadia University, Surat, 394350, India
| | - Jitendra Singh Yadav
- Shri Ram Murti Smarak College of Engineering and Technology (Pharmacy), Bareilly, Uttar Pradesh, 243202, India
| | - Purnima Tripathi
- Anangpuria Institute of Pharmaceutical Sciences, Alampur, Ballabgarh, Faridabad, Haryana, 121004, India
| |
Collapse
|
50
|
Liao T, Chen X, Qiu F, Zhang X, Wu F, Zhao Z, Xu M, Chen M, Shen JW, Shen Q, Ji J. Regulation of cancer-associated fibroblasts for enhanced cancer immunotherapy using advanced functional nanomedicines: an updated review. J Nanobiotechnology 2025; 23:166. [PMID: 40038745 PMCID: PMC11877876 DOI: 10.1186/s12951-025-03217-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The tumor microenvironment (TME) is a complex and dynamic ecosystem that plays a critical role in cancer progression. It comprises various cell types, including immune cells, tumor cells, and stromal cells. Among these, cancer-associated fibroblasts (CAFs) represent a heterogeneous population with diverse origins, phenotypes, and functions. Activated CAFs secrete multiple factors that promote tumor growth, migration, angiogenesis, and contribute to chemoresistance. Additionally, CAFs secrete extracellular matrix (ECM) components, such as collagen, which form a physical barrier that hinders the penetration of chemotherapeutic and immunotherapeutic agents. This ECM also influences immune cell infiltration, impeding their ability to effectively target tumor cells. As a result, modulating the activity of CAFs has emerged as a promising strategy to enhance the efficacy of tumor immunotherapy. Nano-delivery systems, constructed from various nanomaterials with high targeting specificity and biocompatibility, offer a compelling approach to deliver therapeutic agents or immunomodulatory factors directly to CAFs. This modulation can alter CAF function, reduce their tumor-promoting effects, and thereby improve the outcomes of immunotherapy. This review provides an in-depth exploration of the origins, functions, and interactions of CAFs within the TME, particularly in the context of immune suppression. Furthermore, it discusses the potential applications of functional nanocarrifers in modulating CAFs and enhancing the effectiveness of tumor immunotherapy, highlighting the significant progress and potential of nanotechnology in this area.
Collapse
Affiliation(s)
- Tingting Liao
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China
| | - Xiaoxiao Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Fengkai Qiu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Xinyu Zhang
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Fazong Wu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Ming Xu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| | - Jia-Wei Shen
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Qiying Shen
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Jiansong Ji
- School of Pharmacy, College of Pharmacy, Hangzhou Normal University, 2318 Yuhangtang Road, Hangzhou, 310015, Zhejiang, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, 289 Kuocang Road, Lishui, 323000, China.
- Department of Radiology, Lishui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|