1
|
Liu S, Ren Z, Yan M, Ye W, Hu Y. Strategies to enhance the penetration of nanomedicine in solid tumors. Biomaterials 2025; 321:123315. [PMID: 40185056 DOI: 10.1016/j.biomaterials.2025.123315] [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: 12/22/2024] [Revised: 03/16/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Nanomedicine was previously regarded as a promising solution in the battle against cancer. Over the past few decades, extensive research has been conducted to exploit nanomedicine for overcoming tumors. Unfortunately, despite these efforts, nanomedicine has not yet demonstrated its ability to cure tumors, and the research on nanomedicine has reached a bottleneck. For a significant period of time, drug delivery strategies have primarily focused on targeting nanomedicine delivery to tumors while neglecting its redistribution within solid tumors. The uneven distribution of nanomedicine within solid tumors results in limited therapeutic effects on most tumor cells and significantly hampers the efficiency of drug delivery and treatment outcomes. Therefore, this review discusses the challenges faced by nanomedicine in penetrating solid tumors and provides an overview of current nanotechnology strategies (alleviating penetration resistance, size regulation, tumor cell transport, and nanomotors) that facilitate enhanced penetration of nanomedicine into solid tumors. Additionally, we discussed the potential role of nanobionics in promoting effective penetration of nanomedicine.
Collapse
Affiliation(s)
- Sen Liu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Zhendong Ren
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Manqi Yan
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Wei Ye
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Yong Hu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
2
|
Huang Y, Chen L, Chen Y, Zhou S, Xie X, Xie J, Yu M, Chen J. High-density lipoprotein-based nanoplatform reprograms tumor microenvironment and enhances chemotherapy against pancreatic ductal adenocarcinoma. Biomaterials 2025; 318:123147. [PMID: 39908877 DOI: 10.1016/j.biomaterials.2025.123147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/25/2024] [Accepted: 01/26/2025] [Indexed: 02/07/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly aggressive, with limited success in traditional therapies due to the fibrotic, immunosuppressive, pro-metastatic tumor microenvironment (TME), which collectively impede the drug accumulation and accelerate the tumor progression. In this work, we developed a PDAC-customized nutrient-mimicking reconstituted high-density lipoprotein (rHDL) capable of efficiently co-encapsulate versatile TME regulating cannabidiol and cytotoxic gemcitabine to simultaneously reprogram TME while suppressing PDAC progression. Specifically, a small-sized, nutrient-like rHDL was constructed to realize deep PDAC parenchyma penetration and efficient intra-tumoral uptake. Next, natural herbal compound cannabidiol was screened and incorporated into rHDL to regulate TME via attenuating fibrosis, reliving immunosuppression and mitigating metastatic tendency. At last, gemcitabine, the PDAC gold standard first-line therapy was co-delivered by the PDAC-customized rHDL to overcome drug resistance and amplify its PDAC suppression. Our findings demonstrate the feasibility of an integrated multi-stage TME regulation strategy for improved PDAC therapy, and might represent a modality in promoting chemotherapy against PDAC.
Collapse
Affiliation(s)
- Yukun Huang
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China; Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liang Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yu Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Songlei Zhou
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaoying Xie
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Jing Xie
- Department of Minimally Invasive Therapy Center, Fudan University Shanghai Cancer Center, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Minghua Yu
- Fudan University Clinical Research Center for Cell-based Immunotherapy & Department of Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Shanghai, 201399, China
| | - Jun Chen
- Shanghai Pudong Hospital & Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
3
|
Sonzini S, England RM, Kapustin AN, Moss JI, Sutton D, Smith A, Sharma S, Siouve E, Mazza M, Ravn P, Puri S, Ashford M. HER2-targeted star polymer conjugates for improved tumor distribution and efficacy. J Control Release 2025; 382:113654. [PMID: 40122243 DOI: 10.1016/j.jconrel.2025.113654] [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: 11/19/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Actively targeted nanoparticle systems have the potential to improve delivery to tumors over untargeted systems however the design rules to achieve this have not been fully elucidated. A HER2-targeted polymer drug delivery system composed of a 32-arm star polymer (SD) conjugated with the TOP1 inhibitor molecule SN-38, with a trastuzumab antigen binding fragment (HER2-Fab), has been used to target cancer cells overexpressing this receptor. The HER2-Fab was attached to the SD at two different densities (average of 1 or 3 Fabs per star polymer) and compared to the native star polymer without Fab. In vitro experimentation showed that both the targeted star polymers (HER2-SDs) had better binding and uptake in HER2-positive cell lines (SK-BR3 and HEK293) compared to the non-targeted SD. In vivo biodistribution studies showed enhanced accumulation of HER2-targeted SDs in tumors, but not normal tissues, particularly at the later (96 h post-dose) timepoint. The HER2-SDs demonstrated increased localization with tumor cells rather than in stromal regions, greater penetration into the tumor core and a more homogenous distribution in the tumor section than the untargeted SD. The targeted star polymer conjugated to SN-38 was tested for anti-tumor activity in a HER2-positive gastric cancer xenograft in mice and showed significantly greater efficacy compared to untargeted SDs.
Collapse
Affiliation(s)
- Silvia Sonzini
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Richard M England
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Alexander N Kapustin
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jennifer I Moss
- OTD Small Molecules, Early Oncology R&D, AstraZeneca, Cambridge, UK
| | | | | | - Soumya Sharma
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Elise Siouve
- Biologic Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Mariarosa Mazza
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Peter Ravn
- Biologic Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Sanya Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Marianne Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| |
Collapse
|
4
|
Martin JD, Toh K, Martin MR, Chen P, Wang C, Igarashi K, Mpekris F, Stylianopoulos T, Stuber MD, Kataoka K, Cabral H. Bone marrow vessels are hyperpermeable to macromolecules and nanoscale medicine in a size-dependent manner. J Control Release 2025; 382:113669. [PMID: 40158811 DOI: 10.1016/j.jconrel.2025.113669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/22/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Bone marrow (BM) has roles in health and disease, so systemically administered nanocarriers (NCs) targeting or avoiding BM are desirable. While the hydrodynamic diameter of NCs can be tuned to target or avoid various organs, the size dependence of extravasation from BM vessels is unknown. To clarify the size dependence of passive transvascular transport in the BM, we performed vessel permeability measurements in murine calvaria using confocal fluorescent microscopy with fluorescently labeled dextrans, albumin, and polymeric micelles as model probes. Unexpectedly, we found the permeability of BM vessels to macromolecules decreases with increasing hydrodynamic diameter between 4 nm and 32 nm. We modeled this permeability data with mathematical models to predict an effective pore size for sinusoids of 47 nm and non-sinusoids of 37 nm, with estimated maximum pore sizes of 61 nm and 53 nm, respectively. Finally, we tested these model predictions by demonstrating that the extravasation of 70 nm polymeric micelles, which are larger than the estimated maximum pore size, is hindered relative to 30 nm polymeric micelles. These results establish design criteria for controlling NC hydrodynamic diameter towards modulating delivery to BM.
Collapse
Affiliation(s)
- John D Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, Japan
| | - Margaret R Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Pengwen Chen
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Chenyu Wang
- Process Systems and Operations Research Laboratory, Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA
| | - Kazunori Igarashi
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Matthew D Stuber
- Process Systems and Operations Research Laboratory, Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA.
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, Kawasaki, Kanagawa, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan.
| |
Collapse
|
5
|
Lantzberg B, Zeyn Y, Forster R, Jian L, Schauenburg D, Hieber C, Nuhn L, Zhou T, Silva MJSA, Koynov K, Jiang HL, Kuan SL, Bros M, Opatz T, Weil T. Glycogen-inspired trimannosylated serum albumin nanocarriers for targeted delivery of toll-like receptor 7/8 agonists to immune cells and liver. J Control Release 2025; 382:113705. [PMID: 40199455 DOI: 10.1016/j.jconrel.2025.113705] [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: 09/20/2024] [Revised: 03/20/2025] [Accepted: 04/04/2025] [Indexed: 04/10/2025]
Abstract
Nanocarriers can improve the therapeutic efficiency of small molecule immunomodulators or inhibitors, which is important for immunotherapy of liver diseases or cancer. Macromolecular protein carriers, such as human serum albumin (HSA), could provide better penetration compared to large nanoparticles (>50 nm) but are hampered by systemic biodistribution. To overcome these limitations, inspired by the natural glycogen structure, we have designed the HSA nanocarrier (<40 nm) consisting of multiple trimannose (TM) ligands attached to the protein surface, to target mannose or dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) receptors in immune cells or immunological organs such as the liver. Capitalizing on the chemical reactivity of different amino acids present in HSA, we have incorporated multiple copies of a cargo relevant for immunotherapy, i.e. the toll-like receptor (TLR) 7/8 agonist. The resulting TM-HSA conjugates exhibit excellent and specific uptake ex vivo in various immune cells and liver-specific uptake in vivo, opening access to protein nanocarriers with rapid and efficient in vivo targeting with great potential for immune-related diseases.
Collapse
Affiliation(s)
- Bellinda Lantzberg
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Yanira Zeyn
- Department of Dermatology, University Medical Center of Johannes Gutenberg-University Mainz, Obere Zahlbacher Straße 63, 55131 Mainz, Germany
| | - Robert Forster
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Lin Jian
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Dominik Schauenburg
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Christoph Hieber
- Department of Dermatology, University Medical Center of Johannes Gutenberg-University Mainz, Obere Zahlbacher Straße 63, 55131 Mainz, Germany
| | - Lutz Nuhn
- Faculty for Chemistry and Pharmaceutics, Julius-Maximilians-University Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | - Tianjiao Zhou
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany; State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, PR China
| | - Maria J S A Silva
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Kaloian Koynov
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210009, PR China
| | - Seah Ling Kuan
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Matthias Bros
- Department of Dermatology, University Medical Center of Johannes Gutenberg-University Mainz, Obere Zahlbacher Straße 63, 55131 Mainz, Germany.
| | - Till Opatz
- Department of Chemistry, Johannes Gutenberg University Mainz, Duesbergweg 10-14, 55128 Mainz, Germany.
| | - Tanja Weil
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| |
Collapse
|
6
|
Qu Y, Guo B, Zhao S, Sun J, Cao J, Xia M, Zhong Z, Meng F. Photothermal treatment of prostate tumor with micellar indocyanine green and napabucasin to co-ablate cancer cells and cancer stem cells. J Control Release 2025; 382:113704. [PMID: 40194599 DOI: 10.1016/j.jconrel.2025.113704] [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: 12/28/2024] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Advanced prostate cancer is hassled by relapse and metastasis that are closely associated with cancer stem cells (CSCs). Here, we present micellar indocyanine green and napabucasin (mICG-Nap) that co-ablates cancer cells and CSCs via photothermal therapy (PTT) for the treatment of prostate tumor. mICG-Nap with stable loading of both drugs and favorable size effectively reduced CSC population in RM1-PSMA murine prostate cancer cells and inhibited tumor spheroid formation. mICG-Nap showed an enhanced photothermal effect compared with free ICG and eliminated tumor spheroids under near-infrared (NIR) irradiation. The efficacy of mICG-Nap was further enhanced by decorating with Acupa ligand, which targets to RM1-PSMA cells and tumors via PSMA receptor. The enhanced tumor cell uptake of Acupa-mICG-Nap led to significant survival benefits in both subcutaneous RM1-PSMA tumor models and postoperative models. The tumor analyses demonstrated clear downregulation of CSC-related biomarkers such as OCT4, SOX2, CD133 and pSTAT3 as well as PSMA by Acupa-mICG-Nap. Rational formulated micellar indocyanine green and napabucasin plus NIR appears as an appealing strategy to co-ablate cancer cells and CSCs with rapid tumor de-bulking yet no recurrence.
Collapse
Affiliation(s)
- Yanyi Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Beibei Guo
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Songsong Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Juan Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Jun Cao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Mingyu Xia
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, PR China.
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
7
|
Zhang BD, Chen X, Su JY, Zhuo SH, Zhao L, Wu JJ, Li WH, Wang TY, Liu L, Yang T, Yang LJ, Zhao YF, Li YM. Rationally designed anti-autophagy nanosystems for reversing the immunosuppressive network in the tumor environment. Nanomedicine (Lond) 2025:1-12. [PMID: 40401367 DOI: 10.1080/17435889.2025.2508133] [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: 09/27/2024] [Accepted: 05/15/2025] [Indexed: 05/23/2025] Open
Abstract
AIMS To develop a nano-immunotherapy system combining autophagy inhibition and innate immune activation to reverse the immunosuppressive tumor microenvironment (TME) in pancreatic ductal adenocarcinoma (PDAC). MATERIALS & METHODS The pH-responsive polymer PC7A was utilized to co-deliver the autophagy inhibitor chloroquine (CQ) and the STING agonist cyclic diguanylate (CDG), forming the CQCP nanosystem. In vitro and in vivo experiments evaluated autophagy inhibition, MHC-I expression, dendritic cell activation, tumor infiltration of lymphocytes, and survival in PDAC-bearing mice. RESULTS CQCP enhanced MHC-I expression on PDAC cells by 2.1-fold (p < 0.001) and increased activated dendritic cells (CD86+/CD40+) by 3.5-fold (p < 0.01) in the TME. Tumor-infiltrating CD8+ T cells rose by 42.6% (p < 0.001), and systemic immune activation in peripheral lymphoid tissues was observed. CQCP achieved an 86% survival rate in tumor-bearing mice, significantly outperforming monotherapies or free drug combinations. CONCLUSIONS The CQCP system synergistically reverses PDAC immunosuppression by restoring antigen presentation and activating innate immunity. This dual-targeted strategy demonstrates robust antitumor efficacy and offers a promising immunotherapy approach for PDAC.
Collapse
Affiliation(s)
- Bo-Dou Zhang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Xi Chen
- Zhili College, Tsinghua University, Beijing, China
| | - Jing-Yun Su
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Shao-Hua Zhuo
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Lang Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Jun-Jun Wu
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Tian-Yang Wang
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Ling Liu
- Peking University First Hospital, Beijing, China
| | - Tao Yang
- Key Laboratory of Digestive Disease & Organ Transplantation in Shanxi Province, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li-Jun Yang
- Key Laboratory of Digestive Disease & Organ Transplantation in Shanxi Province, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yu-Fen Zhao
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
- Beijing Institute for Brain Disorders, Beijing, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| |
Collapse
|
8
|
Yang J, Zeng Z, Liu Y, Li Y, Xu X. Developing bioinspired delivery systems for enhanced tumor penetration of macromolecular drugs. J Control Release 2025; 383:113845. [PMID: 40379215 DOI: 10.1016/j.jconrel.2025.113845] [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: 11/14/2024] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Macromolecular drugs, such as proteins and nucleic acids, play a pivotal role in treating refractory diseases and hold significant promise in the growing pharmaceutical market. However, without efficient delivery systems, macromolecular drugs are highly susceptible to rapid biodegradation or systemic clearance, underscoring the need for advanced delivery strategies for clinical translation. A major challenge lies in their limited tissue penetration due to large molecular weight and size, which has recently garnered significant attention as it often leads to therapeutic failure or the emergence of resistance. In this review, we first outline the biological barriers limiting macromolecular tissue penetration, then explore the inherent permeation mechanisms of biomacromolecules in biological systems. We then highlight delivery strategies aimed at enhancing the tissue penetration of macromolecular therapeutics, with a particular focus on tissue-adaptive and tissue-remodeling delivery platforms. Finally, we provide a concise perspective on future research directions in deep tissue penetration for biomacromolecules. This review offers a comprehensive summary of recent advancements and presents critical insights into optimizing the therapeutic efficacy of macromolecular drugs.
Collapse
Affiliation(s)
- Jin Yang
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China; State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, Hunan 410082, China
| | - Zenan Zeng
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yiming Liu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China
| | - Yachao Li
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China; State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, Hunan 410082, China
| | - Xianghui Xu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan 410082, China; State Key Laboratory of Chemo and Biosensing, Hunan University, Changsha, Hunan 410082, China.
| |
Collapse
|
9
|
Pan Y, Wang X, Zhou X, Chen H, Zou Y. High α-lipoic acid-loaded hollow mesoporous prussian blue nanozymes for targeted therapy of nasopharyngeal carcinoma in mice. J Biomater Appl 2025; 39:1177-1187. [PMID: 39905972 DOI: 10.1177/08853282251318514] [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: 02/06/2025]
Abstract
This study successfully constructs a tumor-targeting α-lipoic acid-loaded hollow mesoporous prussian blue nanozyme (AHPRzyme) for targeted therapy of nasopharyngeal carcinoma in mice. In these nanozymes, Arg-Gly-Asp (RGD) acts as a targeting ligand, enabling effective targeting of tumor cells. Additionally, AHPRzyme exhibits multiple anti-tumor mechanisms: ① The prussian blue nanozymes in AHPRzyme have catalase (CAT) activity, which decomposes H2O2 in human nasopharyngeal carcinoma CEN2 cells into non-toxic H2O, reducing H2O2 levels and minimizing damage to normal cells. The released O2 helps alleviate the hypoxic environment of the tumor, inhibiting lactate production due to hypoxia and consequently suppressing tumor growth. ② The prussian blue nanozymes also have peroxidase (POD) activity, which catalyzes H2O2 in tumor cells to generate ·OH, a reactive oxygen species, leading to tumor cell apoptosis. ③ The α-lipoic acid structure in AHPRzyme contains disulfide bonds that react with GSH, depleting excess glutathione (GSH) in tumor cells, disrupting the oxidative stress balance within the cells, and making them more sensitive to reactive oxygen species, thereby increasing tumor cell apoptosis. In summary, AHPRzyme can inhibit tumor cell growth and promote tumor cell apoptosis by improving the tumor microenvironment, achieving the goal of anti-nasopharyngeal carcinoma therapy.
Collapse
Affiliation(s)
- Ya Pan
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xiaofeng Wang
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Xuejun Zhou
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Haipeng Chen
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Yuxia Zou
- Department of Otolaryngology, Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
10
|
Dar AI, Randhawa S, Verma M, Saini TC, Acharya A. Debugging the dynamics of protein corona: Formation, composition, challenges, and applications at the nano-bio interface. Adv Colloid Interface Sci 2025; 342:103535. [PMID: 40319752 DOI: 10.1016/j.cis.2025.103535] [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: 05/15/2024] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
The intricate interplay between nanomaterials and the biological molecules has garnered considerable interest in understanding the dynamics of protein corona formation at the nano-bio interface. This review provides an in-depth exploration of protein-nanoparticle interactions, elucidating their structural dynamics and thermodynamics at the nano-Bio interface and further on emphasizing its formation, composition, challenges, and applications in the biomedical and nanotechnological domains, such as drug delivery, theranostics, and the translational medicine. We delve the nuanced mechanisms governing protein corona formation on nanoparticle surfaces, highlighting the influence of nanoparticle and biological factors, and review the impact of corona formation on the biological identity and functionality of nanoparticles. Notably, emerging applications of artificial intelligence and machine learning have begun to revolutionize this field, enabling accurate prediction of corona composition and related biological outcomes. These tools not only enhance efficiency over traditional experimental methods but also help decode complex protein-nanoparticle interaction patterns, offering new insights into corona-driven cellular responses and disease diagnostics. Additionally, it discusses recent advancements in the field of protein corona, particularly in translational nanomedicine and associated applications entailing current and future strategies which are aimed at mitigating the adverse effects of protein-nanoparticle interactions at the biological interface, for tailoring the protein coronas by engineering of the nanomaterials. This comprehensive assessment from chemical, technological, and biological aspects serves as a guiding beacon for the development of future nanomedicine, enabling the more effective emulation of the biological milieu and the design of protein-NP systems for enhanced biomedical applications.
Collapse
Affiliation(s)
- Aqib Iqbal Dar
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shiwani Randhawa
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mohini Verma
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Trilok Chand Saini
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
11
|
Yu Q, Zhou Y, Zhang Q, Li J, Yan S, Xu J, Li C, Zhou X, Sun Y. NIR-II imaging guided on-site size variable clustered nanosystem to potentiate sonodynamic therapy in deep-seated tumors. Biomaterials 2025; 322:123381. [PMID: 40319680 DOI: 10.1016/j.biomaterials.2025.123381] [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: 01/23/2025] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
The insufficient enrichment and penetration of sonosensitizers in the tumor site hamper the antitumor efficiency of sonodynamic therapy (SDT). Herein, tumor-acidity and photothermal controlled nanosystems (NTTD), which coloaded a new type of sonosensitizers, Na3TiF6 NPs and second near-infrared (NIR-II) emissive AIEgen (T1), have been developed to achieve highly efficient SDT/photothermal therapy (PTT) in deep-seated tumors. On one hand, NTTD includes ultrasmall Na3TiF6 NPs with increased oxygen vacancies, narrow bandgap (2.82 eV) and preferrable absorption capability of H2O and O2 molecules, guaranteeing powerful generation of reactive oxygen species (ROS) under US stimulation. On the other hand, with the assistance of the acidic/photothermal responses and deep penetrated NIR-II fluorescence imaging (up to 7 mm), NTTD undergo in situ "two-step" size transformation to achieve enhanced retention and penetration of sonosensitizers in tumor area with high spatiotemporal controllability. In 4T1 tumor model, compared to passive targeting participated NTT group, NTTD elongated the tumor retention time to 60 h and revealed enhanced imaging signal (∼2.4 fold). Further photoirradiation of NTTD assisted ∼4.5-fold enhancement of penetration ability. SDT/PTT synergies have evoked significant ROS generation and tumor inhibition rate of 75.2 % in vivo. This study presents an innovative strategy to exploit novel nano-sonosensitizers with precisely improved tumor accumulation and penetration.
Collapse
Affiliation(s)
- Qi Yu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China.
| | - Yujing Zhou
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Qin Zhang
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Juan Li
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Shan Yan
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Jie Xu
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Cao Li
- Key Laboratory of Fermentation Engineering (Ministry of Education), National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, 430068, China
| | - Xiaobo Zhou
- School of Public Health, Nantong Key Laboratory of Public Health and Medical Analysis, Nantong University, Nantong, 226019, China.
| | - Yao Sun
- State Key Laboratory of Green Pesticides, International Joint Research Center for Intelligent Biosensor Technology and Health, College of Chemistry, Central China Normal University, Wuhan, 430079, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China.
| |
Collapse
|
12
|
Sun J, Li HL, Zhou WJ, Ma ZX, Huang XP, Li C. Current status and recent progress of nanomaterials in transcatheter arterial chemoembolization therapy for hepatocellular carcinoma. World J Clin Oncol 2025; 16:104435. [PMID: 40290691 PMCID: PMC12019268 DOI: 10.5306/wjco.v16.i4.104435] [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: 12/23/2024] [Revised: 02/06/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most common cancers worldwide. Transcatheter arterial chemoembolization has become a common treatment modality for some patients with unresectable advanced HCC. Since the introduction of nanomaterials in 1974, their use in various fields has evolved rapidly. In medical applications, nanomaterials can serve as carriers for the delivery of chemotherapeutic drugs to tumour tissues. Additionally, nanomaterials have potential for in vivo tumour imaging. This article covers the properties and uses of several kinds of nanomaterials, focusing on their use in transcatheter arterial chemoembolization for HCC treatment. This paper also discusses the limitations currently associated with the use of nanomaterials.
Collapse
Affiliation(s)
- Jia Sun
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Hai-Liang Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Wen-Jun Zhou
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Zeng-Xin Ma
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Xiao-Pei Huang
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| | - Cheng Li
- Department of Hepatobiliary Pancreatic Hernia Surgery, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou 510317, Guangdong Province, China
| |
Collapse
|
13
|
Lin S, Zhang L, Cui H, Wang Y, Zheng Y, Hu J, Li M, Wang W, Zhang S, Zhou K, Chen Q, Lan X, Zhao Y. Pharmacokinetics modulation in solid tumors through thrombin-embedded nanomedicine. J Nanobiotechnology 2025; 23:268. [PMID: 40186247 PMCID: PMC11969998 DOI: 10.1186/s12951-025-03302-4] [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: 12/16/2024] [Accepted: 03/07/2025] [Indexed: 04/07/2025] Open
Abstract
The development of anti-tumor nanomedicines grapples with the critical challenge of achieving sustained retention and massive intratumoral distributions of chemotherapeutics. Herein, we attempted multifaceted prodrug nanomedicine with precise spatiotemporal responsiveness, integrating dual prodrugs-redox-responsive SN38 and pH-responsive thrombin and ensuring drug release coinciding with the striking tumor acidity and reductive stress, while its spatial selectivity is directed by the overexpression of integrins on cancerous cells. Most importantly, the thrombin component induces vascular occlusion within tumors, leading to normalization of the elevated interstitial fluid pressure and promoting accumulation of chemotherapeutic agents. This approach not only facilitates the massive intratumoral distribution of the nanomedicine but also ensures sustained retention of SN38 within the tumor microenvironment, thereby augmenting the cytotoxic potencies. Of note, the advanced mass spectrum mapping technology unprecedentedly validated the successful activation of the SN38 prodrug and massive distribution throughout the solid tumors for thrombin-containing nanomedicine, in stark to apparent entrapment in tumor vasculature and stroma for the conventional thrombin-free nanomedicine. Hence, the multifunctionalities of our proposed dual prodrug nanomedicine is underscored by its ability to actively target cancerous cells, induce vasculature occlusion, and orchestrate a controlled release of chemotherapeutic agents.
Collapse
Affiliation(s)
- Sheng Lin
- The State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Liuwei Zhang
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Hongyan Cui
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Yue Wang
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Yang Zheng
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
- Department of Clinical Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Jianhua Hu
- The State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200433, China
| | - Mingzhu Li
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
- Department of Integrated Traditional Chinese and Western Medicine Medical Oncology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Wentao Wang
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Shijia Zhang
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
| | - Kehui Zhou
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China
| | - Qixian Chen
- Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzheng, 518120, China.
| | - Xiabin Lan
- Department of Thyroid Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China.
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, 310022, Zhejiang, China.
| | - Yan Zhao
- Department of Gastric Surgery, Cancer Hospital of Dalian University of Technology, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
- Department of Gastric Surgery, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China.
- Provincial Key Laboratory of Interdisciplinary Medical Engineering for Gastrointestinal Carcinoma, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, China.
| |
Collapse
|
14
|
Wei Y, Wang M. Tumor-Targeting Theranostic Polymers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:7928-7945. [PMID: 40118780 DOI: 10.1021/acs.langmuir.4c04978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Theranostic polymers have emerged as a versatile platform in cancer nanomedicine, integrating therapeutic and imaging functionalities to overcome challenges in oncology. Featuring diverse architectures such as linear polymers, dendrimers, star-like polymers, and bottle-brush polymers, these systems enable tumor-targeted drug delivery, real-time imaging, and controlled release. Recent advances in stimuli-responsive designs and biomimetic strategies have improved their specificity, stability, and adaptability, outperforming conventional nanocarriers. This review summarizes the design, synthesis, and biomedical applications of theranostic polymers, focusing on their potential to address tumor heterogeneity and biological barriers. The challenges of biocompatibility, immunogenicity, and clinical translation are discussed, with a perspective toward future developments in precision medicine and imaging-guided cancer therapy.
Collapse
Affiliation(s)
- Ying Wei
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Mingfeng Wang
- School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| |
Collapse
|
15
|
Peixoto D, Ravasco JM, Blanco-Fernandez B, Veiga F, Concheiro A, Conde J, Paiva-Santos AC, Alvarez-Lorenzo C. Enzyme-responsive vitamin D-based micelles for paclitaxel-controlled delivery and synergistic pancreatic cancer therapy. Mater Today Bio 2025; 31:101555. [PMID: 40026626 PMCID: PMC11869029 DOI: 10.1016/j.mtbio.2025.101555] [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: 09/16/2024] [Revised: 12/20/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most feared diseases worldwide owing to its poor prognosis, negligible therapeutic advances, and high mortality. Herein, multifunctional enzyme-responsive micelles for the controlled delivery of paclitaxel (PTX) were prepared to circumvent its current clinical challenges. Accordingly, two enzyme-responsive structural units composed of Vitamin D3 (VD3) conjugated with polyethylene glycol of different molecular weights (600 Da and 2000 Da) were synthesized and characterized using different analytical methods. By applying the solvent evaporation method, these bioactive structural units self-assembled into sub-100 nm VD3 micelles with minimal batch-to-batch variation, monomodal particle size distribution, and high encapsulation efficiency. The enzyme-triggered disassembly of PTX-loaded VD3 micelles was demonstrated by release studies in the presence of a high esterase content typically featured by PDAC cells. PTX-loaded VD3 micelles also exhibited prominent cell internalization and induced a considerable cytotoxic synergistic effect against human PDAC cells (BxPC-3 cells) in 2D and 3D cell culture models compared with free PTX. The PTX-loaded VD3 micelles were hemocompatible and stable after long-term storage in the presence of biorelevant media, and showed higher efficiency to inhibit the tumor growth compared to the approved clinical nanoformulation (Abraxane®) in an in ovo tumor model. The findings reported here indicate that VD3S-PEG micelles may have a promising role in PDAC therapy, since VD3 could act not only as a hydrophobic core of the micelles but also as a therapeutic agent that provides synergetic therapeutic effects with the encapsulated PTX.
Collapse
Affiliation(s)
- Diana Peixoto
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| | - João M. Ravasco
- Comprehensive Health Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169, Lisboa, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649, Lisbon, Portugal
| | - Barbara Blanco-Fernandez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| | - Francisco Veiga
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| | - João Conde
- Comprehensive Health Research Centre, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade Nova de Lisboa, 1169, Lisboa, Portugal
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, 1649, Lisbon, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
- REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Faculty of Pharmacy, iMATUS and Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, 15782, Santiago, Spain
| |
Collapse
|
16
|
Fukumitsu N, Matsumoto Y, Chen L, Sugawara Y, Fujisawa N, Niiyama E, Ouchi S, Oe E, Saito T, Ebara M. Development of Layer-by-Layer Magnetic Nanoparticles for Application to Radiotherapy of Pancreatic Cancer. Molecules 2025; 30:1382. [PMID: 40142157 PMCID: PMC11946117 DOI: 10.3390/molecules30061382] [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: 12/27/2024] [Revised: 02/07/2025] [Accepted: 03/18/2025] [Indexed: 03/28/2025] Open
Abstract
Pancreatic cancer is among the deadliest malignancies, with few treatment options for locally advanced, unresectable cases. Conventional therapies, such as chemoradiotherapy and hyperthermia, show promise but face challenges in improving outcomes. This study introduces a novel drug delivery system using gemcitabine (GEM)-loaded layer-by-layer magnetic nanoparticles (LBL MNPs) combined with alternating magnetic field (AMF) application and X-ray irradiation to enhance therapeutic efficacy. LBL MNPs were synthesized using optimized layering techniques to achieve superior drug loading and controlled release. Human pancreatic cancer cells (PANC-1) were treated with LBL MNPs alone, with AMF-induced hyperthermia, and in combination with X-rays. The results demonstrate that the 7-layer LBL MNPs exhibited optimal cytotoxicity, significantly reducing cell viability at concentrations of 30 µg/mL and higher. Combining 7-layer LBL MNPs with AMF increased cell death in a time- and concentration-dependent manner, achieving up to 98% inhibition of cell proliferation. The addition of X-rays to the regimen demonstrated a strong synergistic effect, resulting in a 13-fold increase in cell death compared to controls. These findings highlight the potential of this integrated approach to improve outcomes in patients with pancreatic cancer.
Collapse
Affiliation(s)
| | - Yoshitaka Matsumoto
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan;
| | - Lili Chen
- Smart Polymers Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Tsukuba 305-0044, Japan; (L.C.); (N.F.); (S.O.); (E.O.); (M.E.)
| | - Yu Sugawara
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan;
| | - Nanami Fujisawa
- Smart Polymers Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Tsukuba 305-0044, Japan; (L.C.); (N.F.); (S.O.); (E.O.); (M.E.)
| | - Eri Niiyama
- Smart Polymers Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Tsukuba 305-0044, Japan; (L.C.); (N.F.); (S.O.); (E.O.); (M.E.)
| | - Sosuke Ouchi
- Smart Polymers Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Tsukuba 305-0044, Japan; (L.C.); (N.F.); (S.O.); (E.O.); (M.E.)
| | - Emiho Oe
- Smart Polymers Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Tsukuba 305-0044, Japan; (L.C.); (N.F.); (S.O.); (E.O.); (M.E.)
| | - Takashi Saito
- Department of Radiation Oncology, Clinical Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
- Proton Medical Research Center, University of Tsukuba Hospital, Tsukuba 305-8576, Japan;
| | - Mitsuhiro Ebara
- Smart Polymers Group, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, Tsukuba 305-0044, Japan; (L.C.); (N.F.); (S.O.); (E.O.); (M.E.)
| |
Collapse
|
17
|
Tang L, Li N, Yang Z, Lin Y, Gao G, Lin Q, Wang Y. Targeted Nanoclusters for Intratracheal Delivery in Intraoperative Imaging of Lung Adenocarcinoma. Int J Nanomedicine 2025; 20:3575-3594. [PMID: 40125441 PMCID: PMC11930283 DOI: 10.2147/ijn.s509009] [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: 12/06/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Background Computed tomography (CT) is widely used all over the world, and the detection rate of early lung adenocarcinoma is increasing. Minimally invasive thoracic surgery (MITS) has emerged as the preferred surgical approach for lung adenocarcinoma, but identifying small lung adenocarcinomas is difficult. Therefore, there is a need for a non-invasive, convenient and efficient way to localize lung adenocarcinomas. Materials and Methods A targeted gold nanocluster for intraoperative fluorescence imaging by intratracheal delivery has been designed. Au-GSH-anti Napsin A nanoclusters (NapA-AuNCs) were synthesized, and their physicochemical properties and optical properties were characterized. Target effect, cytotoxicity and fluorescence time curve of NapA-AuNCs, were tested in vivo and in vitro, and intratracheal delivery was also carried. Results NapA-AuNCs targeting lung adenocarcinoma with red fluorescence and good mucus penetration were synthesized, which had the targeting property of A549 and lung adenocarcinoma tissue, and also had very low toxicity to normal lung epithelial cells and organs. Intracheal delivery involves faster imaging of lung adenocarcinoma and less accumulation of other organs than intravenous administration. Conclusion NapA-AuNCs targeting lung adenocarcinoma were successfully conjugated, and intratracheal delivery is a safe, effective for lung adenocarcinoma intraoperative localization.
Collapse
Affiliation(s)
- Lu Tang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Ning Li
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Zhe Yang
- Shandong Laboratory of Advanced Materials and Green Manufacturing at Yantai, Yanatai, Shandong, People’s Republic of China
| | - Yangliu Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Ge Gao
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Quan Lin
- State Key Laboratory of Supramolecular Structure and Material, College of Chemistry, Jilin University, Changchun, Jilin, People’s Republic of China
| | - Yue Wang
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
18
|
Della Pelle G, Markelc B, Bozic T, Šribar J, Krizaj I, Zagar Soderznik K, Hudoklin S, Kreft ME, Urbančič I, Kisovec M, Podobnik M, Kostevšek N. Red Blood Cell Membrane Vesicles for siRNA Delivery: A Biocompatible Carrier With Passive Tumor Targeting and Prolonged Plasma Residency. Int J Nanomedicine 2025; 20:3269-3301. [PMID: 40109366 PMCID: PMC11921803 DOI: 10.2147/ijn.s504644] [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: 11/04/2024] [Accepted: 02/04/2025] [Indexed: 03/22/2025] Open
Abstract
Background Despite many advances in gene therapy, the delivery of small interfering RNAs is still challenging. Erythrocytes are the most abundant cells in the human body, and their membrane possesses unique features. From them, erythrocytes membrane vesicles can be generated, employable as nano drug delivery system with prolonged blood residence and high biocompatibility. Methods Human erythrocyte ghosts were extruded in the presence of siRNA, and the objects were termed EMVs (erythrocyte membrane vesicles). An ultracentrifugation-based method was applied to select only the densest EMVs, ie, those containing siRNA. We evaluated their activity in vitro in B16F10 cells expressing fluorescent tdTomato and in vivo in B16F10 tumor-bearing mice after a single injection. Results The EMVs had a negative zeta potential, a particle size of 170 nm and excellent colloidal stability after one month of storage. With 0.3 nM siRNA, more than 75% gene knockdown was achieved in vitro, and 80% was achieved in vivo, at 2 days PI at 2.5 mg/kg. EMVs mostly accumulate around blood vessels in the lungs, brain and tumor. tdTomato fluorescence steadily decreased in tumor areas with higher EMVs concentration, which indicates efficient gene knockdown. Approximately 2% of the initial dose of EMVs was still present in the plasma after 2 days. Conclusion The entire production process of the purified siRNA-EMVs took approximately 4 hours. The erythrocyte marker CD47 offered protection against macrophage recognition in the spleen and in the blood. The excellent biocompatibility and pharmacokinetic properties of these materials make them promising platforms for future improvements, ie, active targeting and codelivery with conventional chemotherapeutics.
Collapse
Affiliation(s)
- Giulia Della Pelle
- Department for Nanostructured Materials, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
- Jožef Stefan International Postgraduate School, Ljubljana, 1000, Slovenia
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Jernej Šribar
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Igor Krizaj
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | | | - Samo Hudoklin
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Mateja Erdani Kreft
- Institute of Cell Biology, Faculty of Medicine, University of Ljubljana, Ljubljana, 1000, Slovenia
| | - Iztok Urbančič
- Laboratory of Biophysics, Condensed Matter Physics Department, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| | - Matic Kisovec
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, 1000, Slovenia
| | - Marjetka Podobnik
- Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Ljubljana, 1000, Slovenia
| | - Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, Ljubljana, 1000, Slovenia
| |
Collapse
|
19
|
Li Y, Sun H, Cao D, Guo Y, Wu D, Yang M, Wang H, Shao X, Li Y, Liang Y. Overcoming Biological Barriers in Cancer Therapy: Cell Membrane-Based Nanocarrier Strategies for Precision Delivery. Int J Nanomedicine 2025; 20:3113-3145. [PMID: 40098719 PMCID: PMC11913051 DOI: 10.2147/ijn.s497510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/04/2025] [Indexed: 03/19/2025] Open
Abstract
Given the unique capabilities of natural cell membranes, such as prolonged blood circulation and homotypic targeting, extensive research has been devoted to developing cell membrane-inspired nanocarriers for cancer therapy, while most focused on overcoming one or a few biological barriers. In fact, the journey of nanosystems from systemic circulation to tumor cells involves intricate processes, encompassing blood circulation, tissue accumulation, cancer cell targeting, endocytosis, endosomal escape, intracellular trafficking to target sites, and therapeutic action, all of which pose limitations to their clinical translation. This underscores the necessity of meticulously considering these biological barriers in the design of cell membrane-mimetic nanocarriers. In this review, we delineate the functions and applications of diverse types of cell membranes in nanocarrier systems. We elaborate on the biological hurdles encountered at each stage of the biomimetic nanoparticle's odyssey to the target, and comprehensively discuss the obstacles imposed by the tumor microenvironment for precise delivery. Subsequently, we systematically review contemporary cell membrane-based strategies aimed at overcoming these multi-level biological barriers, encompassing hybrid cell membrane (HCM) camouflage, tumor microenvironment remodeling, endosomal/lysosomal escape, multidrug resistance (MDR) reversal, optimization of nanoparticle physicochemical properties, and so on. Finally, we outline potential strategies to accelerate the development of cell membrane-inspired precision nanocarriers and discuss the challenges that must be addressed to enhance their clinical applicability. This review serves as a guide for refining the study of cell membrane-mimetic nanosystems in surmounting in vivo delivery barriers, thereby significantly contributing to advancing the development and application of cell membrane-based nanoparticles in cancer delivery.
Collapse
Affiliation(s)
- Yuping Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dianchao Cao
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Dongyang Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Menghao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hongming Wang
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Xiaowei Shao
- Binzhou Inspection and Testing Center, Binzhou, ShanDong, 256600, People’s Republic of China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| |
Collapse
|
20
|
Rosales-Barrios C, González-Sánchez ZI, Zuliani A, Jiménez-Vacas JM, Luque RM, Pozo D, Khiar N. PSMA-targeted delivery of docetaxel in prostate cancer using small-sized PDA-based micellar nanovectors. J Control Release 2025; 379:890-905. [PMID: 39864631 DOI: 10.1016/j.jconrel.2025.01.062] [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: 08/05/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
In this study, we present the first comparative analysis of active and passive drug delivery systems for docetaxel (DTX) in prostate cancer using supramolecular self-assembled micellar nanovectors. Specifically, we developed two novel micelles based on polydiacetylenic amphiphiles (PDA) for passive and active targeting. The active targeting micelles were designed with a prostate-specific membrane antigen (PSMA) ligand, ACUPA, to facilitate recognition by PSMA-positive cancer cells. These PDA-based micelles feature a well-defined structure with a hydrophobic PDA core and a surface functionalized with PEG, and for active targeting, ACUPA. Our micelles demonstrated excellent encapsulation capacity, significantly improving DTX solubility in water, a crucial factor for clinical drug use. In vitro studies confirmed the safety and cytotoxic profiles of both systems, with ACUPA-functionalized micelles showing notable internalization into PSMA-positive LNCaP cells, mediated through the PSMA-ACUPA interaction. In vivo imaging revealed preferential accumulation of ACUPA-functionalized nanomicelles in LNCaP xenograft tumors, suggesting enhanced retention via specific ACUPA-PSMA interactions and active uptake by LNCaP cells. Notably, Balb/c-Foxn1nu/nu early in vivo studies showed a marked reduction in tumor volume and tumor expression levels of proliferation, cell cycle progression, cell survival and anti-apoptotic markers with DTX-loaded micelles functionalized with ACUPA compared to those without ACUPA. Overall, our studies collect initial evidence regarding the feasibility of supramolecular self-assembly of ACUPA-PDA-based nanomicelles for PSMA-targeted drug chemotherapy delivery developments.
Collapse
Affiliation(s)
- Cristian Rosales-Barrios
- Asymmetric Synthesis and Functional Nanosystems Group (Art&Fun), Institute of Chemical Research (IIQ), CSIC-University of Seville, C/ Américo Vespucio 49, 41092 Seville, Spain
| | - Zaira I González-Sánchez
- Department of Integrative Pathophysiology and Therapies, Andalusian Centre for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-Universidad Pablo de Olavide-Universidad de Sevilla, Av. Americo Vespucio 24, Seville 41092, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology University of Seville, Av. Sánchez Pizjuan s/n, 41009 Seville, Spain; Nanobiology Laboratory, Department of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra (PUCMM), Hwy. Duarte km 1.5, Santiago de los Caballeros, 822, Dominican Republic
| | - Alessio Zuliani
- Asymmetric Synthesis and Functional Nanosystems Group (Art&Fun), Institute of Chemical Research (IIQ), CSIC-University of Seville, C/ Américo Vespucio 49, 41092 Seville, Spain
| | - Juan M Jiménez-Vacas
- Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University Hospital Reina Sofía (HURS), Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av. Menéndez Pidal s/n, Córdoba 14004, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Campus Rabanales, 14004 Córdoba, Spain
| | - Raul M Luque
- Maimónides Institute for Biomedical Research of Córdoba (IMIBIC), University Hospital Reina Sofía (HURS), Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Av. Menéndez Pidal s/n, Córdoba 14004, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Campus Rabanales, 14004 Córdoba, Spain
| | - David Pozo
- Department of Integrative Pathophysiology and Therapies, Andalusian Centre for Molecular Biology and Regenerative Medicine (CABIMER), CSIC-Universidad Pablo de Olavide-Universidad de Sevilla, Av. Americo Vespucio 24, Seville 41092, Spain; Department of Medical Biochemistry, Molecular Biology and Immunology University of Seville, Av. Sánchez Pizjuan s/n, 41009 Seville, Spain
| | - Noureddine Khiar
- Asymmetric Synthesis and Functional Nanosystems Group (Art&Fun), Institute of Chemical Research (IIQ), CSIC-University of Seville, C/ Américo Vespucio 49, 41092 Seville, Spain.
| |
Collapse
|
21
|
Lee S, Kang S, Kim WJ. Targeted Protein Degradation in Cancer Therapy via Hydrophobic Polymer-Tagged Nanoparticles. ACS NANO 2025; 19:7742-7754. [PMID: 39982901 DOI: 10.1021/acsnano.4c12747] [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: 02/23/2025]
Abstract
Targeted protein degradation (TPD) strategies offer a significant advantage over traditional small molecule inhibitors by selectively degrading disease-causing proteins. While small molecules can lead to recurrence and resistance due to compensatory pathway activation, TPD addresses this limitation by promoting protein degradation, thereby reducing the likelihood of recurrence and resistance over the long-term. Despite these benefits, bifunctional TPD molecules face challenges such as low solubility, poor bioavailability, and limited tumor specificity. In this study, we developed polymer-based nanoparticles that combine TPD strategies with nanotechnology through a hydrophobic tagging method. Hydrophobic polymer-tagged nanoparticles facilitate targeted protein degradation by incorporating hydrophobic polymers that mimic hydrophobic residues in misfolded proteins. This system combines degradation and delivery capabilities within a polymer-based platform, inducing protein degradation while improving solubility, stability, and tumor targeting. These nanoparticles consist of a block copolymer composed of an androgen receptor ligand (ARL)-conjugated hydrophobic polylactic acid (PLA) and a hydrophilic polyethylene glycol (PEG), connected by a GSH-cleavable disulfide bond. In aqueous solutions, this block copolymer (ARL-PLA-SS-PEG) forms micelles that degrade in reducible cellular environments. The micelles demonstrated significant in vitro degradation of the target androgen receptor (AR). Furthermore, they achieved substantial tumor accumulation and significantly inhibited tumor growth in a tumor-bearing mouse model. A mechanistic study revealed that the micelle-mediated TPD follows a dual pathway involving both proteasome and autophagosome. This approach has the potential to serve as a universal platform for protein degradation, eliminating the need to develop disease-specific TPD molecules.
Collapse
Affiliation(s)
- Seohee Lee
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Seonwoo Kang
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Won Jong Kim
- Department of Chemistry, POSTECH-CATHOLIC Biomedical Engineering Institute, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
- OmniaMed Co, Ltd, Pohang 37673, Republic of Korea
| |
Collapse
|
22
|
Sivakumar PM, Zarepour A, Akhter S, Perumal G, Khosravi A, Balasekar P, Zarrabi A. Anionic polysaccharides as delivery carriers for cancer therapy and theranostics: An overview of significance. Int J Biol Macromol 2025; 294:139211. [PMID: 39732249 DOI: 10.1016/j.ijbiomac.2024.139211] [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: 11/13/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Recently, cancer therapy has witnessed remarkable advancements with a growing focus on precision medicine and targeted drug delivery strategies. The application of anionic polysaccharides has gained traction in various drug delivery systems. Anionic polysaccharides have emerged as promising delivery carriers in cancer therapy and theranostics, offering numerous advantages such as biocompatibility, low toxicity, and the ability to encapsulate and deliver therapeutic agents to tumor sites with high specificity. This review underscores the significance of anionic polysaccharides as essential components of the evolving landscape of cancer therapy and theranostics. These polymers can be tailored to carry a wide range of therapeutic cargo, including chemotherapeutic agents, nucleic acids, and imaging agents. Their negative charge enables electrostatic interactions with positively charged drugs and facilitates the formation of stable nanoparticles, liposomes, or hydrogels for controlled drug release. Additionally, their hydrophilic nature aids in prolonging circulation time, reducing drug degradation, and minimizing off-target effects. Besides, some of them could act as targeting agents or therapeutic compounds that lead to improved therapeutic performance. This review offers valuable information for researchers, clinicians, and biomedical engineers. It provides insights into the recent progress in the applications of anionic polysaccharide-based delivery platforms in cancer theranostics to transform patient outcomes.
Collapse
Affiliation(s)
- Ponnurengam Malliappan Sivakumar
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam; School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India.
| | - Sohail Akhter
- New Product Development, Global R&D, Sterile ops, TEVA Pharmaceutical Industries Ltd., Aston Ln N, Halton, Preston Brook, Runcorn WA7 3FA, UK.
| | - Govindaraj Perumal
- Department of Biomedical Engineering, School of Dental Medicine, University of Connecticut (UConn) Health, Farmington, CT 06030, USA.
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye
| | - Premkumar Balasekar
- Department of Pharmacology, K.K. College of Pharmacy, Affiliated to The Tamilnadu Dr. M.G.R. Medical University, Gerugambakkam 600128, India
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
23
|
Tang J, Zhang J, Li Y, Hu Y, He D, Ni H, Zhang J, Wu F, Tang Y, Wang S. Interpretable Radiomics Model Predicts Nanomedicine Tumor Accumulation Using Routine Medical Imaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416696. [PMID: 39916575 DOI: 10.1002/adma.202416696] [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: 10/30/2024] [Revised: 12/18/2024] [Indexed: 03/27/2025]
Abstract
Accurately predicting nanomedicine accumulation is critical for guiding patient stratification and optimizing treatment strategies in the context of precision medicine. However, non-invasive prediction of nanomedicine accumulation remains challenging, primarily due to the complexity of identifying relevant imaging features that predict accumulation. Here, a novel non-invasive method is proposed that utilizes standard-of-care medical imaging modalities, including computed tomography and ultrasound, combined with a radiomics-based model to predict nanomedicine accumulation in tumor. The model is validated using a test dataset consisting of seven tumor xenografts in mice and three sizes of gold nanoparticles, achieving an area under the receiver operating characteristic curve of 0.851. The median accumulation levels of tumors predicted as "high accumulators" are 2.69 times greater than those predicted as "low accumulators". Analysis of this machine-learning-driven interpretable radiomics model revealed imaging features that are strongly correlated with dense stroma, a recognized biological barrier to effective nanomedicine delivery. Radiomics-based prediction of tumor accumulation holds promise for stratifying patient and enabling precise tailoring of nanomedicine treatment strategies.
Collapse
Affiliation(s)
- Jiajia Tang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Jie Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yang Li
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yongzhi Hu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Doudou He
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Hao Ni
- Department of Mathematics, University College London, London, WC1H0AY, UK
| | - Jiulou Zhang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Feiyun Wu
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Yuxia Tang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| | - Shouju Wang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210000, China
| |
Collapse
|
24
|
Jiang L, Wu A, Zeng L, Zhou B, Zhao M, Fan M, Jin Z, He Q. A Slimming/Excavating Strategy for Enhanced Intratumoral Penetration of Acid-Disassemblable NO-Releasing Nanomedicines. Adv Healthc Mater 2025; 14:e2404085. [PMID: 39757461 DOI: 10.1002/adhm.202404085] [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: 11/01/2024] [Revised: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Poor tumor penetration is the major predicament of nanomedicines that limits their anticancer efficacy. The dense extracellular matrix (ECM) in the tumor is one of the major barriers against the deep penetration of nanomedicines. In this work, a slimming/excavating strategy is proposed for enhanced intratumoral penetration based on an acid-disassemblable nanomicelles-assembled nanomedicine and the NO-mediated degradation of ECM. The nanomedicine is constructed by cross-linking nanomicelles, which are self-assembled with two kinds of dendrimers containing phenylboronic acid and lactobionic acid, through borate esterification. In the acidic tumor microenvironment, the pH-sensitive borate ester bonds among the nanomicelles are hydrolyzed, triggering the disassembly of nanomedicine (≈150 nm) into small nanomicelles (≈25 nm). In response to the intratumoral over-expressed glutathione (GSH), the NO donor loaded in the nanomicelles produces NO, which mediates the expression of matrix metalloproteinases for the degradation of ECM in the tumor. By collaboration of the disassembling behavior of nanomedicine with the NO-mediated degradation of ECM, the designed nanomedicine can penetrate a long distance in tumors. The proposed slimming/excavating strategy will provide inspiration for overcoming the challenge of nanomedicines in tumor penetration.
Collapse
Affiliation(s)
- Lingdong Jiang
- College of Pharmacy, Shenzhen Technology University, Shenzhen, 518118, China
| | - Anbang Wu
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Lingting Zeng
- Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bin Zhou
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Min Zhao
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Mingjian Fan
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Zhaokui Jin
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, Guangdong, 518060, China
| | - Qianjun He
- Shanghai Key Laboratory of Hydrogen Science & Center of Hydrogen Science, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
25
|
Ansari M, Kulkarni YA, Singh K. Ocular polymeric nanomicelles for the posterior eye segment in the management of retinoblastoma: formulation, optimization, in vitro and ex vivo evaluations. Pharm Dev Technol 2025; 30:246-258. [PMID: 39985152 DOI: 10.1080/10837450.2025.2469321] [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: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
The existing study focuses on the development, optimization, and evaluation of sorafenib-loaded polymeric nanomicelles for posterior segment delivery in treating retinoblastoma. The formulation involved adjusting various process and product parameters to create effective drug-loaded polymeric nanomicelles. The particle size, PDI, and zeta potential of optimized formulation were found to be 65.52 ± 1.18 nm, 0.14 ± 0.01, and -3.26 ± 0.66 mV, respectively. The entrapment efficiency and drug release were estimated to be 98.84% ± 0.001 and 99.99% in 6 h, respectively. Additionally, the optimized formulation demonstrated acceptable outcomes for solid-state analysis, osmolality, pH, residual solvent, and morphological properties. After 8 h, the ex vivo transcleral permeation and scleral deposition were 629.05 ± 124.11 ng/cm2 and 4.10 ± 0.54 µg, respectively. Y-79 (human retinoblastoma) cell line study using standard drug, test drug, and optimized formulation revealed anticancer potential at all time points (6, 12, 18, and 24 h) with comparable IC50 values. Furthermore, the optimized formulation exhibited no toxicity on the ARPE-19 (human retinal pigmented epithelium) cell line over 24 h. The optimized formulation was non-irritating to the eye (HET-CAM) and remained stable for 6 months. Thus, drug delivery to the posterior eye segment for the treatment of retinoblastoma appears to be possible with the help of established technology.
Collapse
Affiliation(s)
- Mudassir Ansari
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed to be University, Mumbai, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed to be University, Mumbai, India
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed to be University, Mumbai, India
| |
Collapse
|
26
|
Wang R, Zhang Y, Guo Y, Zeng W, Li J, Wu J, Li N, Zhu A, Li J, Di L, Cao P. Plant-derived nanovesicles: Promising therapeutics and drug delivery nanoplatforms for brain disorders. FUNDAMENTAL RESEARCH 2025; 5:830-850. [PMID: 40242551 PMCID: PMC11997602 DOI: 10.1016/j.fmre.2023.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/17/2023] [Accepted: 09/10/2023] [Indexed: 04/18/2025] Open
Abstract
Plant-derived nanovesicles (PDNVs), including plant extracellular vesicles (EVs) and plant exosome-like nanovesicles (ELNs), are natural nano-sized membranous vesicles containing bioactive molecules. PDNVs consist of a bilayer of lipids that can effectively encapsulate hydrophilic and lipophilic drugs, improving drug stability and solubility as well as providing increased bioavailability, reduced systemic toxicity, and enhanced target accumulation. Bioengineering strategies can also be exploited to modify the PDNVs to achieve precise targeting, controlled drug release, and massive production. Meanwhile, they are capable of crossing the blood-brain barrier (BBB) to transport the cargo to the lesion sites without harboring human pathogens, making them excellent therapeutic agents and drug delivery nanoplatform candidates for brain diseases. Herein, this article provides an initial exposition on the fundamental characteristics of PDNVs, including biogenesis, uptake process, isolation, purification, characterization methods, and source. Additionally, it sheds light on the investigation of PDNVs' utilization in brain diseases while also presenting novel perspectives on the obstacles and clinical advancements associated with PDNVs.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yingjie Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Yumiao Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Wei Zeng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jinge Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jie Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Nengjin Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Anran Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Jiale Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial TCM Engineering Technology Research Center of High Efficient Drug Delivery System, Nanjing 210023, China
| | - Peng Cao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Provincial Medical Innovation Center, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
| |
Collapse
|
27
|
Roszkowski S, Durczyńska Z, Szablewska S. Targeted nanodelivery systems for personalized cancer therapy. Rep Pract Oncol Radiother 2025; 29:776-788. [PMID: 40104662 PMCID: PMC11912883 DOI: 10.5603/rpor.103524] [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: 11/27/2023] [Accepted: 11/12/2024] [Indexed: 03/20/2025] Open
Abstract
Conventional cancer therapies such as chemotherapy face challenges such as poor tumor targeting, systemic toxicity, and drug resistance. Nanotechnology offers solutions through advanced drug delivery systems that preferentially accumulate in tumors while avoiding healthy tissues. Recent innovations have enabled the optimization of engineered nanocarriers for extended circulation and tumor localization via both passive and active targeting mechanisms. Passive accumulation exploits the leaky vasculature of tumors, whereas active strategies use ligands to selectively bind cancer cell receptors. Multifunctional nanoparticles also allow the combination of imaging, multiple therapeutic modalities and on-demand drug release within a single platform. Overall, precisely tailored nanotherapeutics that leverage unique pathophysiological traits of malignancies provide opportunities to overcome the limitations of traditional treatment regimens. This emerging field promises more effective and personalized nanomedicine approaches to detect and treat cancer. The key aspects highlighted in this review include the biological barriers associated with nanoparticles, rational design principles to optimize nanocarrier pharmacokinetics and tumor uptake, passive and active targeting strategies, multifunctionality, and reversal of multidrug resistance.
Collapse
Affiliation(s)
- Szymon Roszkowski
- Division of Biochemistry and Biogerontology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Zofia Durczyńska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| | - Sylwia Szablewska
- Department of Oncology, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz,
Poland
| |
Collapse
|
28
|
Pradhan A, Biswal S, Bhal S, Biswal BK, Kundu CN, Subuddhi U, Pati A, Hassan PA, Patel S. Amphiphilic Poly(ethylene glycol)-Cholesterol Conjugate: Stable Micellar Formulation for Efficient Loading and Effective Intracellular Delivery of Curcumin. ACS APPLIED BIO MATERIALS 2025; 8:1418-1436. [PMID: 39907519 DOI: 10.1021/acsabm.4c01657] [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: 02/06/2025]
Abstract
A biodegradable and biocompatible micellar-based drug delivery system was developed using amphiphilic methoxy-poly(ethylene glycol)-cholesterol (C1) and poly(ethylene glycol)-S-S-cholesterol (C2) conjugates and applied to the tumoral release of the water-insoluble drug curcumin. These synthesized surfactants C1 and C2 were found to form stable micelles (CMC ∼ 6 μM) and an average hydrodynamic size of around 20-25 nm. The curcumin-encapsulated C1 micelle was formulated by a solvent evaporation method. A very high drug encapsulation efficiency (EE) of ∼88% and a drug loading (DL) capacity of ∼9% were determined for both the micelles. From the reduced rate of curcumin degradation and differential scanning calorimetry (DSC) analysis, the stability of the curcumin-loaded C1 micelle was found to be higher than that of the unloaded micelle, which confirmed a more compact structural arrangement in the presence of hydrophobic curcumin. A pH-sensitive release of curcumin (faster release with decrease in pH) was observed for the curcumin-loaded C1 micelle, attributed to the diffusion and relaxation/erosion of micellar aggregates. To achieve reduction environment-sensitive drug release, a disulfide (S-S) chemical linkage-incorporated mPEG-cholesterol conjugate (C2) was synthesized, which was found to show glutathione-responsive faster release of curcumin. The in vitro experiments carried out in SCC9 oral cancer cell lines showed that the blank C1 and C2 micelles were noncytotoxic at lower concentrations (<50 μM), while curcumin-loaded C1 and C2 micelles inhibited the proliferation and promoted the apoptosis. An increased in vitro cytotoxicity was observed for curcumin-loaded micelles compared to that of curcumin itself, demonstrating a better cell penetration efficacy of the micelle. These results were further supplemented by the in vivo anticancer analysis of the curcumin-loaded C1 and C2 micellar formulations using the mice xenograft model. Notably, curcumin-loaded C2 micelles showed a significantly stronger apoptotic effect in xenograft mice compared to curcumin-loaded C1 micelles, indicating the GSH environment-sensitive drug release and improved bioavailability. In conclusion, the mPEG-cholesterol C1 and C2 micellar system with the advantages of small size, high encapsulation efficiency, high drug loading, simple preparing technique, biocompatibility, and good in vitro and in vivo performance may have the potential to be used as a drug carrier for sustained and stimuli-responsive release of the hydrophobic drug curcumin.
Collapse
Affiliation(s)
- Aiswarya Pradhan
- Department of Chemistry, National Institute of Technology, Rourkela 769 008, India
| | - Stuti Biswal
- Department of Life Sciences, National Institute of Technology, Rourkela 769 008, India
| | - Subhasmita Bhal
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, India
| | - Bijesh K Biswal
- Department of Life Sciences, National Institute of Technology, Rourkela 769 008, India
| | - Chanakya Nath Kundu
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, India
| | - Usharani Subuddhi
- Department of Chemistry, National Institute of Technology, Rourkela 769 008, India
| | - Anita Pati
- School of Applied Sciences, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar 751024, India
| | - P A Hassan
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Sabita Patel
- Department of Chemistry, National Institute of Technology, Rourkela 769 008, India
| |
Collapse
|
29
|
Shiomi K, Hayashi K, Ishii H, Kamei T, Shimanouchi T, Nakamura H, Ichikawa S. Phase-separated cationic giant unilamellar vesicles as templates for the polymerization of tetraethyl orthosilicate (TEOS). BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184403. [PMID: 39626823 DOI: 10.1016/j.bbamem.2024.184403] [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: 04/08/2024] [Revised: 11/22/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
Unlike homogeneous liposomes, phase-separated liposomes have the potential to be attractive soft materials because they exhibit different properties for each phase. In this study, phase separation was observed when liposomes were prepared using 1,2-dioleoyloxy-3-trimethylammonium propane chloride (DOTAP), 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC), and cholesterol. The pH of the DOTAP-rich phase was evaluated using a coumarin derivative, and measurements showed that DOTAP molecules accumulated hydroxyl ions (OH-) in the DOTAP-rich phase. Such accumulation of OH- was not observed when homogeneous DSPC liposomes were used. The difference in local concentration of OH- in each phase was applied to prepare hollow silica particles with large pores. The OH- promotes the polymerization of tetraethyl orthosilicate (TEOS). Therefore, TEOS polymerized preferentially in the DOTAP-rich phase, whereas a silica membrane barely formed in the DSPC-rich phase.
Collapse
Affiliation(s)
- Kohei Shiomi
- Master's Program in Agro-Bioresources Science and Technology, Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan; Department of Materials Science and Chemical Engineering, Faculty of Advanced Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan
| | - Keita Hayashi
- Department of Chemical Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan.
| | - Haruyuki Ishii
- Department of Sustainable Environmental Engineering, Graduate School of Sciences and Technology for Innovation, Yamaguchi University, 2-16-1, Tokiwadai, Ube 755-8611, Japan
| | - Toshiyuki Kamei
- Department of Chemical Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan
| | - Toshinori Shimanouchi
- Division of Environmental Science, Graduate School of Environmental and Life Science, Okayama University, Kita-ku, Okayama 700-8530, Japan
| | - Hidemi Nakamura
- Department of Chemical Engineering, National Institute of Technology, Nara College, 22 Yata-cho, Yamatokoriyama, Nara 639-1080, Japan
| | - Sosaku Ichikawa
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| |
Collapse
|
30
|
Ma T, Tran TB, Lin E, Hunt S, Haveman R, Castro K, Lu J. Size-transformable nanotherapeutics for cancer therapy. Acta Pharm Sin B 2025; 15:834-851. [PMID: 40177555 PMCID: PMC11959941 DOI: 10.1016/j.apsb.2024.11.012] [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: 07/28/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 04/05/2025] Open
Abstract
The size of nanodrugs plays a crucial role in shaping their chemical and physical characteristics, consequently influencing their therapeutic and diagnostic interactions within biological systems. The optimal size of nanomedicines, whether small or large, offers distinct advantages in disease treatment, creating a dilemma in the selection process. Addressing this challenge, size-transformable nanodrugs have surfaced as a promising solution, as they can be tailored to entail the benefits associated with both small and large nanoparticles. In this review, various strategies are summarized for constructing size-transformable nanosystems with a focus on nanotherapeutic applications in the field of biomedicine. Particularly we highlight recent research developments in cancer therapy. This review aims to inspire researchers to further develop various toolboxes for fabricating size-transformable nanomedicines for improved intervention against diverse human diseases.
Collapse
Affiliation(s)
- Teng Ma
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, the University of Arizona, Tucson 85721, AZ, USA
| | - Tuyen Ba Tran
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, the University of Arizona, Tucson 85721, AZ, USA
| | - Ethan Lin
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, the University of Arizona, Tucson 85721, AZ, USA
| | - Stephanie Hunt
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, the University of Arizona, Tucson 85721, AZ, USA
| | - Riley Haveman
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, the University of Arizona, Tucson 85721, AZ, USA
| | - Kylie Castro
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, the University of Arizona, Tucson 85721, AZ, USA
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, the University of Arizona, Tucson 85721, AZ, USA
- Clinical and Translational Oncology Program, the University of Arizona Cancer Center, Tucson 85721, AZ, USA
- BIO5 Institute, the University of Arizona, Tucson 85721, AZ, USA
- Southwest Environmental Health Sciences Center, the University of Arizona, Tucson 85721, AZ, USA
| |
Collapse
|
31
|
Salmasi Z, Kamali H, Rezaee H, Nazeran F, Jafari Z, Eisvand F, Teymouri M, Khordad E, Mosafer J. Simultaneous therapeutic and diagnostic applications of magnetic PLGA nanoparticles loaded with doxorubicin in rabbit. Drug Deliv Transl Res 2025; 15:770-785. [PMID: 39215953 DOI: 10.1007/s13346-024-01693-9] [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] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
In this study, DOX (Doxorubicin) and Fe3O4 magnetic nanocrystals (SPIONs (Superparamagnetic iron oxide nanocrystals)) were encapsulated in the PLGA-PEG: poly(lactide-co-glycolide)-b-poly(ethylene glycol) nanoparticles for theranostic purposes. The final prepared formulation which is called NPs (Nanoparticles) exhibited a particle size with a mean diameter of ~ 209 nm and a sufficient saturation magnetization value of 1.65 emu/g. The NPs showed faster DOX release at pH 5.5 compared to pH 7.4. Also, the cytotoxicity effect of NPs increased compared to Free-DOX alone in C6 glioma cancer cells. For in vivo investigations, the 2.2 Kg rabbits were injected with NPs formulations via a central articular anterior vein in their ears. Furthermore, the images of rabbit organs were depicted via MR (Magnetic resonance) and fluorescent imaging techniques. A negative contrast (dark signal) was observed in T2 (Relaxation Time) weighted MR images of IV (Intravenously)-injected rabbits with NPs compared to the control ones. The organ's florescent images of NPs-injected rabbits showed a high density of red color related to the accumulation of DOX in liver and kidney organs. These data showed that the NPs have no cytotoxicity effect on the heart. Also, the results of histopathological tests of different organs showed that the groups receiving NPs and Free-DOX were almost similar and no significant difference was seen, except for the cardiac tissue in which the pathological effects of NPs were significantly less than the Free-DOX. Additionally, pharmacokinetic studies were also conducted at the sera and whole bloods of IV-injected rabbits with NPs and Free-DOX. The pharmacokinetic parameters showed that NPs could enhance the DOX retention in the serum compared to the Free-DOX. Altogether, we aimed to produce a powerful delivery nanosystem for its potential in dual therapeutic and diagnostic applications which are called theranostic agents.
Collapse
Affiliation(s)
- Zahra Salmasi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Kamali
- Targeted Drug Delivery Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanieh Rezaee
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faezeh Nazeran
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Jafari
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Frarhad Eisvand
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Manouchehr Teymouri
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, North Khorasan, Iran
| | - Elnaz Khordad
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Jafar Mosafer
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran.
- Health Sciences Research Center, Torbat Heydariyeh University of Medical Sciences, P.O. Box 9516915169, Torbat Heydariyeh, Iran.
| |
Collapse
|
32
|
Austria E, Bilek M, Varamini P, Akhavan B. Breaking biological barriers: Engineering polymeric nanoparticles for cancer therapy. NANO TODAY 2025; 60:102552. [DOI: 10.1016/j.nantod.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
Dai X, Yin Y, Wang C, Xu H. Hyaluronic acid regulated facile synthesis of size-tunable multifunctional nanomedicine for effective cancer therapy. Int J Biol Macromol 2025; 288:138668. [PMID: 39667478 DOI: 10.1016/j.ijbiomac.2024.138668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
The complex and heterogeneous nature of cancer necessitates the development of innovative multifunctional nanomedicines (MN). Hyaluronic acid (HA) is a functional carbohydrate polysaccharide that is widely used in various biomedical fields. In this study, we employed HA as a stabilizer and regulator for the synthesis of a size-tunable nanomedicine comprising ferric ions, doxorubicin, and epigallocatechin gallate (EGCG), referred to as HDE-MN, for cancer therapy. A change in the HA ratio can yield HDE-MNs with sizes varying from ~20 nm to over 100 nm. Modified HA can respond to hyaluronidase (HAase) to provide controllable pH/HAase dual-responsive drug release for improved cancer therapy. Moreover, HA can mediate the targeted delivery of HDE-MNs both in vitro and in vivo to cancer cells. In addition, HDE-MNs reversed multidrug resistance owing to the incorporation of EGCG, inducing ferroptosis due to the involvement of ferric ions. More importantly, HDE-MNs have a photothermal conversion effect, enabling photothermal therapy, photothermally enhanced drug release, and ferroptosis, which collectively contribute to significantly improved cancer therapy. Therefore, the HDE-MNs with laser irradiation achieved full ablation of the in vivo tumors. Together with its good biocompatibility, HDE-MNs may be promising for effective cancer therapy.
Collapse
Affiliation(s)
- Xiuliang Dai
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, China
| | - Yina Yin
- Obstetrics and Gynecology Department, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Hongbin Xu
- Obstetrics and Gynecology Department, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China.
| |
Collapse
|
34
|
Han X, Petrova V, Song Y, Cheng YT, Jiang X, Zhou H, Hu C, Chen DS, Yong HJ, Kim HW, Zhang B, Barkai O, Jain A, Renthal W, Lirk P, Woolf CJ, Shi J. Lipid nanoparticle delivery of siRNA to dorsal root ganglion neurons to treat pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.633455. [PMID: 39896578 PMCID: PMC11785206 DOI: 10.1101/2025.01.23.633455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Sensory neurons within the dorsal root ganglion (DRG) are the primary trigger of pain, relaying activity about noxious stimuli from the periphery to the central nervous system; however, targeting DRG neurons for pain management has remained a clinical challenge. Here, we demonstrate the use of lipid nanoparticles (LNPs) for effective intrathecal delivery of small interfering RNA (siRNA) to DRG neurons, achieving potent silencing of the transient receptor potential vanilloid 1 (TRPV1) ion channel that is predominantly expressed in nociceptor sensory neurons. This leads to a reversible interruption of heat-, capsaicin-, and inflammation-induced nociceptive conduction, as observed by behavioral outputs. Our work provides a proof-of-concept for intrathecal siRNA therapy as a novel and selective analgesic modality.
Collapse
|
35
|
Zhang M, Yang Y, Xu Y, Wang J, Li S. Iodinated Copper-Cysteamine Nanoparticles as Radiosensitizers for Tumor Radiotherapy. Pharmaceutics 2025; 17:149. [PMID: 40006516 PMCID: PMC11858929 DOI: 10.3390/pharmaceutics17020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Radiotherapy is a widely applied first-line clinical treatment modality of cancer. Copper-cysteamine (Cu-Cy) nanoparticles represent a new type of photosensitizer that demonstrates significant anti-tumor potential by X-ray-induced photodynamic therapy. Iodide is a high-Z element with superior X-ray absorption ability and has the β-decay radiotherapeutic nuclide, 131I, which emits Cherenkov light. In this study we aimed to investigate the X-ray-induced photodynamic therapy potential of iodinated Cu-Cy (Cu-Cy-I) nanoparticles and also explore the local treatment efficacy of 131I-labeled Cu-Cy-I ([131I]Cu-Cy-I) nanoparticles. Methods: The synthesis of [131I]Cu-Cy-I nanoparticles was performed with [131I]I- anions. The in vitro radiobiological effects on tumor cells incubated with Cu-Cy-I nanoparticles by X-ray irradiation were investigated. The in vivo tumor growth-inhibitory effects of the combination of Cu-Cy-I nanoparticles with X-ray radiotherapy and [131I]Cu-Cy-I nanoparticles were evaluated with 4T1 tumor-xenografted mice. Results: The in vitro experiment results indicated that the X-ray irradiation with the presence of Cu-Cy-I nanoparticles produced a higher intracellular reactive oxygen species (ROS) level and more DNA damage of 4T1 cells and showed a stronger tumor cell killing ability compared to X-ray irradiation alone. The in vivo experimental results with 4T1 breast carcinoma-bearing mice showed that the combination of an intratumoral injection of Cu-Cy-I nanoparticles and X-ray radiotherapy enhanced the tumor growth-inhibitory effect and prolonged the mice's lives. Conclusions: Cu-Cy-I nanoparticles have good potential as new radiosensitizers to enhance the efficacy of external X-ray radiotherapy. However, the efficacy of local treatment with [131I]Cu-Cy-I nanoparticles at a low 131I dose was not verified. The effective synthesis of smaller sizes of nanoparticles is necessary for further investigation of the radiotherapy potential of [131I]Cu-Cy-I nanoparticles.
Collapse
Affiliation(s)
- Miaomiao Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou 215123, China; (M.Z.); (Y.Y.); (Y.X.)
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yu Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou 215123, China; (M.Z.); (Y.Y.); (Y.X.)
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou 215123, China; (M.Z.); (Y.Y.); (Y.X.)
| | - Jie Wang
- Institute for Advanced Materials, School of Material Science and Engineering, Jiangsu University, Zhenjiang 212013, China;
| | - Shihong Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou 215123, China; (M.Z.); (Y.Y.); (Y.X.)
- Department of Clinical Pharmacology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
36
|
Chen MY, Cheng TW, Pan YC, Mou CY, Chiang YW, Lin WC, Hu CMJ, Mou KY. Endotoxin-Free Outer Membrane Vesicles for Safe and Modular Anticancer Immunotherapy. ACS Synth Biol 2025; 14:148-160. [PMID: 39763210 PMCID: PMC11744915 DOI: 10.1021/acssynbio.4c00483] [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: 07/09/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Bacterial outer membrane vesicles (OMVs) have emerged as promising vehicles for anticancer drug delivery due to their inherent tumor tropism, immune-stimulatory properties, and potential for functionalization with therapeutic proteins. Despite their advantages, the high lipopolysaccharide (LPS) endotoxin content in the OMVs raises significant safety and regulatory challenges. In this work, we produce LPS-attenuated and LPS-free OMVs and systematically assess the effects of LPS modification on OMVs' physicochemical characteristics, membrane protein content, immune-stimulatory capacity, tolerability, and anticancer efficacy. Our findings reveal that LPS removal increased the maximal tolerated dose of the OMVs by over 25-fold. When adjusted for comparable safety profiles, LPS-free OMVs exhibit superior anticancer effects compared with wild-type OMVs. Mechanistic investigations indicate that the LPS removal obviates immune cell death caused by LPS and reduces the negatory effects of wild type of OMVs on tumor immune cell infiltrates. We further show the functionality of the LPS-free OMV through the incorporation of an IL-2 variant protein (Neo-2/15). This functionalization augments OMV's ability of the OMV to inhibit tumor growth and promote lymphocyte infiltration into the tumor microenvironment. This study presents a safe and functionalizable OMV with improved translational prospect.
Collapse
Affiliation(s)
- Mei-Yi Chen
- Chemical
Biology and Molecular Biophysics Program, Taiwan International Graduate
Program, Academia Sinica, No. 128, Sec. 2, Academia Rd., Nangang
(Nankang) Dist., Taipei City 115201, Taiwan
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Department
of Chemistry, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Ting-Wei Cheng
- Chemical
Biology and Molecular Biophysics Program, Taiwan International Graduate
Program, Academia Sinica, No. 128, Sec. 2, Academia Rd., Nangang
(Nankang) Dist., Taipei City 115201, Taiwan
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Yi-Chung Pan
- Chemical
Biology and Molecular Biophysics Program, Taiwan International Graduate
Program, Academia Sinica, No. 128, Sec. 2, Academia Rd., Nangang
(Nankang) Dist., Taipei City 115201, Taiwan
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chung-Yuan Mou
- Department
of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yun-Wei Chiang
- Department
of Chemistry, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Wan-Chen Lin
- Chemical
Biology and Molecular Biophysics Program, Taiwan International Graduate
Program, Academia Sinica, No. 128, Sec. 2, Academia Rd., Nangang
(Nankang) Dist., Taipei City 115201, Taiwan
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Che-Ming Jack Hu
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Kurt Yun Mou
- Institute
of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
37
|
Zhang Z, Luo Z, Huang H, Huang Y, Xu J, Liu XY, Zhang W, Li S, Sun J. YAP/TAZ Inhibitor-Based Drug Delivery System for Selective Tumor Accumulation and Cancer Combination Therapy. Biomacromolecules 2025; 26:266-278. [PMID: 39644231 PMCID: PMC11834954 DOI: 10.1021/acs.biomac.4c01076] [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] [Indexed: 12/09/2024]
Abstract
The YES-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are two important transcriptional coactivators that are often aberrantly activated in cancer cells. Their dysregulation promotes cancer development and can confer resistance to anticancer therapies. Therefore, the pharmacological inhibition of YAP/TAZ presents a promising approach for treating tumors with heightened YAP/TAZ activity. However, the clinical use of a known YAP/TAZ inhibitor, niflumic acid (NA), is limited by its poor in vivo half-life. To improve its bioavailability, we developed a series of NA-based prodrug polymers and investigated the impact of NA monomer units on the physicochemical properties of their self-assembled nanoparticles. The optimal pNA polymer was selected as a prodrug micellar nanocarrier to load hydrophobic receptor tyrosine kinase inhibitors (RTKIs) for combination therapy. The nanocarrier selectively accumulated in the tumor and synergistically inhibited tumor growth with the cargo RTKIs, particularly Dasatinib, introducing a nanocombination therapy enhanced breast cancer treatment.
Collapse
Affiliation(s)
- Ziqian Zhang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
| | - Zhangyi Luo
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
| | - Haozhe Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
| | - Yixian Huang
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
| | - Jieni Xu
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
| | - Xian-You Liu
- Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
- Department of Chemistry, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15213, United States
| | - Wei Zhang
- Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
- Department of Chemistry, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15213, United States
| | - Song Li
- Center for Pharmacogenetics, Department of Pharmaceutical Science, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, United States
| | - Jingjing Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68105, United States
| |
Collapse
|
38
|
Konarita K, Kanamori K, Suzuki M, Tokura D, Tanaka S, Honda Y, Nishiyama N, Nomoto T. Poly(vinyl alcohol) potentiating an inert d-amino acid-based drug for boron neutron capture therapy. J Control Release 2025; 377:385-396. [PMID: 39532208 DOI: 10.1016/j.jconrel.2024.11.017] [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: 08/08/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Since the discovery of d-amino acids, they have been considered inactive and have not been used as potent drugs. Here, we report that simple mixing with poly(vinyl alcohol) (PVA) unleashed latent potentials of d-amino acids in boron neutron capture therapy (BNCT). PVA formed boronate esters with seemingly useless boronated d-amino acids and induced tumor-associated amino acid transporter-superselective internalization and prolonged intracellular retention, accomplishing complete cure of tumors. The superselective internalization was achieved by switching the internalization pathway from ineffective pass through the transporter to the transporter-mediated endocytosis. The acidic environment in the endo-/lysosome dissociated the boronate esters and elicited the stealthiness of the drugs, preventing their externalization and prolonging intracellular retention time. In a subcutaneous tumor model, this system accomplished surprisingly high tumor-selective accumulation that could not be achieved by conventional approaches and induced drastic BNCT effects. PVA may be a unique material to unlock potentials of seemingly inert molecules.
Collapse
Affiliation(s)
- Kakeru Konarita
- Department of Life Sciences, Graduate School of Arts and Sciences, the University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Kaito Kanamori
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Minoru Suzuki
- Division of Particle Radiation Oncology, Particle Radiation Oncology Research Center, Institute for Integrated Radiation and Nuclear Science, Kyoto University, 2-1010, Asashiro-nishi, Kumatori-cho, Sennan-gun, Osaka 590-0494, Japan
| | - Daiki Tokura
- Department of Life Sciences, Graduate School of Arts and Sciences, the University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Shota Tanaka
- Department of Life Sciences, Graduate School of Arts and Sciences, the University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| | - Yuto Honda
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan; Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | - Takahiro Nomoto
- Department of Life Sciences, Graduate School of Arts and Sciences, the University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan; Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsutacho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.
| |
Collapse
|
39
|
Wang H, Yang C, Wu T, Fan J, Zhu H, Liu J, Ding B. A Highly Tumor-Permeating DNA Nanoplatform for Efficient Remodeling of Immunosuppressive Tumor Microenvironments. Angew Chem Int Ed Engl 2025; 64:e202412804. [PMID: 39225768 DOI: 10.1002/anie.202412804] [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: 07/08/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/04/2024]
Abstract
The immunosuppressive tumor microenvironment and limited intratumoral permeation have largely constrained the outcome of tumor therapy. Herein, we report a tailored DNA structure-based nanoplatform with striking tumor-penetrating capability for targeted remodeling of the immunosuppressive tumor microenvironment in vivo. In our design, chemo-immunomodulator (gemcitabine) can be precisely grafted on DNA sequences through a reactive oxygen species (ROS)-sensitive linker. After self-assembly, the gemcitabine-grafted DNA structure can site-specifically organize legumain-activatable melittin pro-peptide (promelittin) on each vertex for intratumoral delivery and further function as the template to load photosensitizers (methylene blue) for ROS production. The tailored DNA nanoplatform can achieve targeted accumulation, highly improved intratumoral permeation, and efficient immunogenic cell death of tumor cells by laser irradiation. Finally, the immunosuppressive tumor microenvironment can be successfully remodeled by reducing multi-type immunosuppressive cells and enhancing the infiltration of cytotoxic lymphocytes in the tumor. This rationally developed multifunctional DNA nanoplatform provides a new avenue for the development of tumor therapy.
Collapse
Affiliation(s)
- Hong Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Changping Yang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Tiantian Wu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Pharmaceutical Sciences, Hainan Medical University, Haikou, 570228, China
| | - Jing Fan
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
| | - Hanyin Zhu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianbing Liu
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baoquan Ding
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, National Center for Nanoscience and Technology, Beijing, 100190, China
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, 450001, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
40
|
Cassani M, Fernandes S, Pagliari S, Cavalieri F, Caruso F, Forte G. Unraveling the Role of the Tumor Extracellular Matrix to Inform Nanoparticle Design for Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409898. [PMID: 39629891 PMCID: PMC11727388 DOI: 10.1002/advs.202409898] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Indexed: 01/14/2025]
Abstract
The extracellular matrix (ECM)-and its mechanobiology-regulates key cellular functions that drive tumor growth and development. Accordingly, mechanotherapy is emerging as an effective approach to treat fibrotic diseases such as cancer. Through restoring the ECM to healthy-like conditions, this treatment aims to improve tissue perfusion, facilitating the delivery of chemotherapies. In particular, the manipulation of ECM is gaining interest as a valuable strategy for developing innovative treatments based on nanoparticles (NPs). However, further progress is required; for instance, it is known that the presence of a dense ECM, which hampers the penetration of NPs, primarily impacts the efficacy of nanomedicines. Furthermore, most 2D in vitro studies fail to recapitulate the physiological deposition of matrix components. To address these issues, a comprehensive understanding of the interactions between the ECM and NPs is needed. This review focuses on the main features of the ECM and its complex interplay with NPs. Recent advances in mechanotherapy are discussed and insights are offered into how its combination with nanomedicine can help improve nanomaterials design and advance their clinical translation.
Collapse
Affiliation(s)
- Marco Cassani
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Soraia Fernandes
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
| | - Stefania Pagliari
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Giancarlo Forte
- International Clinical Research CenterSt. Anne's University HospitalBrno60200Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonWC2R 2LSUK
| |
Collapse
|
41
|
Ratnaparkhi MP, Salvankar SS, Tekade AR, Kulkarni GM. Core-Shell Nanoparticles for Pulmonary Drug Delivery. Pharm Nanotechnol 2025; 13:90-116. [PMID: 38265371 DOI: 10.2174/0122117385277725231120043600] [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: 08/17/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 01/25/2024]
Abstract
Nanoscale drug delivery systems have provoked interest for application in various therapies on account of their ability to elevate the intracellular concentration of drugs inside target cells, which leads to an increase in efficacy, a decrease in dose, and dose-associated adverse effects. There are several types of nanoparticles available; however, core-shell nanoparticles outperform bare nanoparticles in terms of their reduced cytotoxicity, high dispersibility and biocompatibility, and improved conjugation with drugs and biomolecules because of better surface characteristics. These nanoparticulate drug delivery systems are used for targeting a number of organs, such as the colon, brain, lung, etc. Pulmonary administration of medicines is a more appealing method as it is a noninvasive route for systemic and locally acting drugs as the pulmonary region has a wide surface area, delicate blood-alveolar barrier, and significant vascularization. A core-shell nano-particulate drug delivery system is more effective in the treatment of various pulmonary disorders. Thus, this review has discussed the potential of several types of core-shell nanoparticles in treating various diseases and synthesis methods of core-shell nanoparticles. The methods for synthesis of core-shell nanoparticles include solid phase reaction, liquid phase reaction, gas phase reaction, mechanical mixing, microwave- assisted synthesis, sono-synthesis, and non-thermal plasma technology. The basic types of core-shell nanoparticles are metallic, magnetic, polymeric, silica, upconversion, and carbon nanomaterial- based core-shell nanoparticles. With this special platform, it is possible to integrate the benefits of both core and shell materials, such as strong serum stability, effective drug loading, adjustable particle size, and immunocompatibility.
Collapse
Affiliation(s)
- Mukesh P Ratnaparkhi
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| | - Shailendra S Salvankar
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| | - Avinash R Tekade
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| | - Gajanan M Kulkarni
- Department of Pharmaceutics, Marathwada Mitra Mandal's College of Pharmacy, Thergaon, Pune, Maharashtra, 411033, India
| |
Collapse
|
42
|
Sivasubramanian PD, Unnikrishnan G, Kolanthai E, Muthuswamy S. Engineered nanoparticle systems: A review on emerging strategies for enhanced cancer therapeutics. NEXT MATERIALS 2025; 6:100405. [DOI: 10.1016/j.nxmate.2024.100405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Martin JD, Mpekris F, Chauhan VP, Martin MR, Walsh ME, Stuber MD, McDonald DM, Yuan F, Stylianopoulos T, Jain RK. Fixation alters the physical properties of tumor tissue that regulate nanomedicine transport. Drug Deliv 2024; 31:2430528. [PMID: 39568143 PMCID: PMC11583359 DOI: 10.1080/10717544.2024.2430528] [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: 07/18/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024] Open
Abstract
To have the desired therapeutic effect, nanomedicines and macromolecular medications must move from the site of injection to the site of action, without having adverse effects. Transvascular transport is a critical step of this navigation, as exemplified by the Enhanced Permeability and Retention (EPR) effect in solid tumors, not found in normal organs. Numerous studies have concluded that passive, diffusion- and convection-based transport predominates over active, cellular mechanisms in this effect. However, recent work using a new approach reevaluated this principle by comparing tumors with or without fixation and concluded the opposite. Here, we address the controversy generated by this new approach by reporting evidence from experimental investigations and computer simulations that separate the contributions of active and passive transport. Our findings indicate that tissue fixation reduces passive transport as well as active transport, indicating the need for new methods to distinguish the relative contributions of passive and active transport.
Collapse
Affiliation(s)
| | - Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Vikash P. Chauhan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Megan E. Walsh
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Matthew D. Stuber
- Process Systems and Operations Research Laboratory, Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA
| | - Donald M. McDonald
- Helen Diller Family Comprehensive Cancer Center, Department of Anatomy, University of California, San Francisco, CA, USA
| | - Fan Yuan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Feng Y, Tang Q, Wang B, Yang Q, Zhang Y, Lei L, Li S. Targeting the tumor microenvironment with biomaterials for enhanced immunotherapeutic efficacy. J Nanobiotechnology 2024; 22:737. [PMID: 39605063 PMCID: PMC11603847 DOI: 10.1186/s12951-024-03005-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024] Open
Abstract
The tumor microenvironment (TME) is a complex system characterized by low oxygen, low pH, high pressure, and numerous growth factors and protein hydrolases that regulate a wide range of biological behaviors in the tumor and have a profound impact on cancer progression. Immunotherapy is an innovative approach to cancer treatment that activates the immune system, resulting in the spontaneous killing of tumor cells. However, the therapeutic efficacy of these clinically approved cancer immunotherapies (e.g., immune checkpoint blocker (ICB) therapies and chimeric antigen receptor (CAR) T-cell therapies) is far from satisfactory due to the presence of immunosuppressive TMEs created in part by tumor hypoxia, acidity, high levels of reactive oxygen species (ROS), and a dense extracellular matrix (ECM). With continuous advances in materials science and drug-delivery technologies, biomaterials hold considerable potential for targeting the TME. This article reviews the advances in biomaterial-based targeting of the TME to advance our current understanding on the role of biomaterials in enhancing tumor immunity. In addition, the strategies for remodeling the TME offer enticing advantages; however, the represent a double-edged sword. In the process of reshaping the TME, the risk of tumor growth, infiltration, and distant metastasis may increase.
Collapse
Affiliation(s)
- Yekai Feng
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinglai Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Bin Wang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yuming Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China.
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
45
|
Chen P, Li S, Nagaoka K, Kakimi K, Kataoka K, Cabral H. Nanoenabled IL-15 Superagonist via Conditionally Stabilized Protein-Protein Interactions Eradicates Solid Tumors by Precise Immunomodulation. J Am Chem Soc 2024; 146:32431-32444. [PMID: 39356776 PMCID: PMC11613988 DOI: 10.1021/jacs.4c08327] [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: 06/20/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Protein complexes are crucial structures that control many biological processes. Harnessing these structures could be valuable for therapeutic therapy. However, their instability and short lifespans need to be addressed for effective use. Here, we propose an innovative approach based on a functional polymeric cloak that coordinately anchors different domains of protein complexes and assembles them into a stabilized nanoformulation. As the polymer-protein association in the cloak is pH sensitive, the nanoformulation also allows targeting the release of the protein complexes to the acidic microenvironment of tumors for aiding their therapeutic performance. Building on this strategy, we developed an IL-15 nanosuperagonist (Nano-SA) by encapsulating the interleukin-15 (IL-15)/IL-15 Receptor α (IL-15Rα) complex (IL-15cx) for fostering synergistic transpresentation in tumors. Upon intravenous administration, Nano-SA stably circulated in the bloodstream, safeguarding the integrity of IL-15cx until reaching the tumor site, where it selectively released the active complex. Thus, Nano-SA significantly amplified the antitumor immune signals while diminishing systemic off-target effects. In murine colon cancer models, Nano-SA achieved potent immunotherapeutic effects, eradicating tumors without adverse side effects. These findings highlight the transformative potential of nanotechnology for advancing protein complex-based therapies.
Collapse
Affiliation(s)
- Pengwen Chen
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Shangwei Li
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Koji Nagaoka
- Department
of Immunotherapeutics, The University of
Tokyo Hospital, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazuhiro Kakimi
- Department
of Immunotherapeutics, The University of
Tokyo Hospital, 7-3-1
Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Innovation
Center of NanoMedicine (iCONM), Kawasaki
Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Horacio Cabral
- Department
of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
46
|
Leng Q, Anand A, Mixson AJ. A Facile and Promising Delivery Platform for siRNA to Solid Tumors. Molecules 2024; 29:5541. [PMID: 39683699 PMCID: PMC11643702 DOI: 10.3390/molecules29235541] [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: 09/17/2024] [Revised: 10/31/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Over 20 years have passed since siRNA was brought to the public's attention. Silencing genes with siRNA has been used for various purposes, from creating pest-resistant plants to treating human diseases. In the last six years, several siRNA therapies have been approved by the FDA, which solely target disease-inducing proteins in the liver. The extrahepatic utility of systemically delivered siRNA has been primarily limited to preclinical studies. While siRNA targeting the liver comprises relatively simple ligand-siRNA conjugates, siRNA treating extrahepatic diseases such as cancer often requires complex carriers. The complexity of these extrahepatic carriers of siRNA reduces the likelihood of their widespread clinical use. In the current report, we initially demonstrated that a linear histidine-lysine (HK) carrier of siRNA, injected intravenously, effectively silenced luciferase expressed by MDA-MB-435 tumors in a mouse model. This non-pegylated linear peptide carrier was easily synthesized compared to the complex cRGD-conjugated pegylated branched peptides our group used previously. Notably, the tumor-targeting component, KHHK, was embedded within the peptide, eliminating the need to conjugate the ligand to the carrier. Moreover, brief bath sonication significantly improved the in vitro and in vivo silencing of these HK siRNA polyplexes. Several other linear peptides containing the -KHHK- sequence were then screened with some carriers of siRNA, silencing 80% of the tumor luciferase marker. Additionally, silencing by these HK siRNA polyplexes was confirmed in a second tumor model. Not only was luciferase activity reduced, but these siRNA polyplexes also reduced the Raf-1 oncogene in the MDA-MB-231 xenografts. These simple-to-synthesize, effective, linear HK peptides are promising siRNA carriers for clinical use.
Collapse
Affiliation(s)
| | | | - A. James Mixson
- Department of Pathology, University of Maryland School of Medicine, 10 S. Pine St., Baltimore, MD 21201, USA; (Q.L.); (A.A.)
| |
Collapse
|
47
|
Silva MLS. Lectin-modified drug delivery systems - Recent applications in the oncology field. Int J Pharm 2024; 665:124685. [PMID: 39260750 DOI: 10.1016/j.ijpharm.2024.124685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Chemotherapy with cytotoxic drugs remains the core treatment for cancer but, due to the difficulty to find general and usable biochemical differences between cancer cells and normal cells, many of these drugs are associated with lack of specificity, resulting in side effects and collateral cytotoxicity that impair patients' adherence to therapy. Novel cancer treatments in which the cytotoxic effect is maximized while adverse effects are reduced can be implemented by developing targeted therapies that exploit the specific features of cancer cells, such as the typical expression of aberrant glycans. Modification of drug delivery systems with lectins is one of the strategies to implement targeted chemotherapies, as lectins are able to specifically recognize and bind to cancer-associated glycans expressed at the surface of cancer cells, guiding the drug treatment towards these cells and not affecting healthy ones. In this paper, recent advances on the development of lectin-modified drug delivery systems for targeted cancer treatments are thoroughly reviewed, with a focus on their properties and performance in diverse applications, as well as their main advantages and limitations. The synthesis and analytical characterization of the cited lectin-modified drug delivery systems is also briefly described. A comparison with free-drug treatments and with antibody-modified drug delivery systems is presented, emphasizing the advantages of lectin-modified drug delivery systems. Main constraints and potential challenges of lectin-modified drug delivery systems, including key difficulties for clinical translation of these systems, and the required developments in this area, are also signalled.
Collapse
Affiliation(s)
- Maria Luísa S Silva
- Centro de Estudos Globais, Universidade Aberta, Rua da Escola Politécnica 147, 1269-001 Lisboa, Portugal.
| |
Collapse
|
48
|
Wang Y, Yu B, Cai M, Li Z, Yang L, Zhang H, Liu W, Wang M. Multifunctional long afterglow nanoparticles with enhanced photothermal effects for in vivo imaging and tumor-targeting therapy. Talanta 2024; 279:126629. [PMID: 39106649 DOI: 10.1016/j.talanta.2024.126629] [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: 05/28/2024] [Revised: 07/17/2024] [Accepted: 07/27/2024] [Indexed: 08/09/2024]
Abstract
Considering the excellent properties such as deep tissue penetration, high signal-to-noise ratio, and in-situ recharge and reactivation, near-infrared luminescence long afterglow nanoparticles show considerable promise for biological application, especially in multifunctional imaging, targeting, and synergistic therapeutic. In this paper, Zn3Ga4GeO11: 0.1 % Cr3+, 1 % Yb3+, 0.1 % Tm3+@Ag-FA (ZGGO@Ag-FA, ZGA-FA) nanoparticles were synthesized by in-situ growth of Ag nanoparticles on the surface of long afterglow nanoparticles, and further modified with folic acid. Through precise adjustments, the luminescent properties of ZnGa2O4 were enhanced and notably boosted the photothermal effect of Ag by leveraging the upconversion emission of ZGGO, with a photothermal conversion efficiency reaching about 59.9 %. The ZGA-FA nanoparticles are ultra-small, measuring less than 50 nm. The modification with folic acid provides the ZGA-FA nanoparticles with excellent tumor-targeting capabilities, demonstrating effective enrichment and retention in tumor tissues, thus enabling long-term imaging and therapy through in vivo re-excitation. Due to its stable photothermal effect, outstanding near-infrared (NIR) afterglow imaging, and red-light charged characteristics, combined with effective tumor-targeting abilities, the therapeutic strategy proposed by this study has significant potential for clinical applications.
Collapse
Affiliation(s)
- Yunjian Wang
- The Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Bin Yu
- The Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China; College of Chemistry and Chemical Engineering, Lanzhou City University, Lanzhou, 730070, PR China
| | - Mingqin Cai
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Zhihui Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China
| | - Lu Yang
- Department of Ophthalmology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, PR China
| | - Hongbi Zhang
- The Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China
| | - Weisheng Liu
- The Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province and State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, PR China.
| | - Min Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
49
|
Ota S, Kosaka H, Honda K, Hoshino K, Goto H, Futagawa M, Takemura Y, Shimizu K. Characterization of Microscopic Structures in Living Tumor by In Vivo Measurement of Magnetic Relaxation Time Distribution of Intratumor Magnetic Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404766. [PMID: 39152928 DOI: 10.1002/adma.202404766] [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: 04/02/2024] [Revised: 07/31/2024] [Indexed: 08/19/2024]
Abstract
Tumor microscopic structure is crucial for determining properties such as cancer type, disease state (key for early diagnosis), and novel therapeutic strategies. Magnetic particle imaging is an early cancer diagnostic tool using magnetic nanoparticles as a tracer, which actualizes cancer theranostics in combination with hyperthermia treatment using the abilities of magnetic nanoparticles as a heat source. This study focuses on the microscopic structures associated with cancer cell distribution, the stromal compartment, and vascularization in different kinds of living tumors by analyzing the intratumor magnetic relaxation response of magnetic nanoparticles injected into the tumors. Furthermore, this study describes a sequential system for the measurement of magnetic relaxation time and analysis of the intratumor structure using nonbiological samples such as viscous fluids and solidified magnetic nanoparticles. Particularly, the fine discriminability achieved by reconstructing a distribution map representing the relationship between magnetic relaxation time and viscosity of medium is demonstrated, based on experimental data with a limited condition number. Observing tumor microscopic structure through the dynamic magnetization response of intratumor magnetic nanoparticles is a low-invasive tool for analyzing tumor tissue without dissection. It holds promise for the advancement of biomedical applications, such as early cancer theranostics, using magnetic nanoparticles.
Collapse
Affiliation(s)
- Satoshi Ota
- Department of Electrical and Electronic Engineering, Shizuoka University, 3-5-1 Johoku, Chuo-ku, Hamamatsu, 432-8561, Japan
| | - Hiroki Kosaka
- Electrical and Electronic Engineering Course, Graduate School of Integrated Science and Technology, Shizuoka University, 3-5-1 Johoku, Chuo-ku, Hamamatsu, 432-8561, Japan
| | - Keita Honda
- Electrical and Electronic Engineering Course, Graduate School of Integrated Science and Technology, Shizuoka University, 3-5-1 Johoku, Chuo-ku, Hamamatsu, 432-8561, Japan
| | - Kota Hoshino
- Electrical and Electronic Engineering Course, Graduate School of Integrated Science and Technology, Shizuoka University, 3-5-1 Johoku, Chuo-ku, Hamamatsu, 432-8561, Japan
| | - Haruki Goto
- Department of Optoelectronics and Nanostructure Science, Graduate School of Science and Technology, Shizuoka University, 3-5-1 Johoku, Chuo-ku, Hamamatsu, 432-8561, Japan
| | - Masato Futagawa
- Department of Electrical and Electronic Engineering, Shizuoka University, 3-5-1 Johoku, Chuo-ku, Hamamatsu, 432-8561, Japan
| | - Yasushi Takemura
- Department of Electrical and Computer Engineering, Yokohama National University, 79-5, Tokiwadai, Hodogaya-ku, Yokohama, 240-8501, Japan
| | - Kosuke Shimizu
- Nanotheranostics Laboratory, Division of Innovative Diagnostic and Therapeutic Research, Institute of Photonics Medicine, Hamamatsu University School of Medicine, 1-20-1, Handayama, Chuo-ku, Hamamatsu, 431-3192, Japan
| |
Collapse
|
50
|
Ma J, Yang H, Tian X, Meng F, Zhai X, Li A, Li C, Wang M, Wang G, Lu C, Bai J. Matrix metalloproteinase 2-responsive dual-drug-loaded self-assembling peptides suppress tumor growth and enhance breast cancer therapy. Bioeng Transl Med 2024; 9:e10702. [PMID: 39545088 PMCID: PMC11558207 DOI: 10.1002/btm2.10702] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/07/2024] [Accepted: 07/04/2024] [Indexed: 11/17/2024] Open
Abstract
Conventional chemotherapeutic agents are limited by their lack of targeting and penetration and their short retention time, and chemotherapy might induce an immune suppressive environment. Peptide self-assembly can result in a specific morphology, and the resulting morphological changes are stimuli responsive to the external environment, which is important for drug permeation and retention of encapsulated chemotherapeutic agents. In this study, a polypeptide (Pep1) containing the peptide sequences PLGLAG and RGD that is responsive to matrix metalloproteinase 2 (MMP-2) was successfully developed. Pep1 underwent a morphological transformation from a spherical structure to aggregates with a high aspect ratio in response to MMP-2 induction. This drug delivery system (DI/Pep1) can transport doxorubicin (DOX) and indomethacin (IND) simultaneously to target tumor cells for subsequent drug release while extending drug retention within tumor cells, which increases immunogenic cell death and facilitates the immunotherapeutic effect of CD4+ T cells. Ultimately, DI/Pep1 attenuated tumor-associated inflammation, enhanced the body's immune response, and inhibited breast cancer growth by combining the actions of DOX and IND. Our research offers an approach to hopefully enhance the effectiveness of cancer treatment.
Collapse
Affiliation(s)
- Jihong Ma
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Haiyan Yang
- Emergency DepartmentYantaishan HospitalYantaiChina
| | - Xue Tian
- School of Basic Medical SciencesShandong Second Medical UniversityWeifangChina
| | - Fanhu Meng
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Xiaoqing Zhai
- School of Clinical MedicineShandong Second Medical UniversityWeifangChina
| | - Aimei Li
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Chuntao Li
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Min Wang
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Guohui Wang
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Chunbo Lu
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| | - Jingkun Bai
- School of Bioscience and TechnologyShandong Second Medical UniversityWeifangChina
| |
Collapse
|