1
|
Chen Y, Fan Z, Xu W, Zhu Z, Tan Z, Hu Y, Kurzina I, Cherdyntseva N, Yang WJ, Wang L. An injectable nanocomposite hydrogel with deep penetration ability for enhanced photothermal and chemotherapy. J Colloid Interface Sci 2025; 685:268-279. [PMID: 39848061 DOI: 10.1016/j.jcis.2025.01.146] [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: 11/22/2024] [Revised: 01/05/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
The excessive extracellular matrix (ECM) in solid tumors significantly inhibits the deep penetration and homogeneous distribution of nanodrugs, which greatly reduces the therapeutic efficacy. In the present work, an injectable polyelectrolyte hydrogel (CD@IPH) containing collagenase and doxorubicin-loaded polyacrylic acid@polyaniline nanoparticles (DOX@NP) were developed for improved photothermal and chemotherapy. The collagenase is released quickly from the polyelectrolyte hydrogel in the first 12 h, effectively degrading ECM and enhancing the deep penetration and evenly distribution of DOX@NP in tumor tissues. Then, the tumor microenvironment-triggered release of DOX from DOX@NP exhibits improved photothermal and chemotherapeutic efficiency. Owing to the excellent photoacoustic and photothermal properties of polyaniline inner cores of DOX@NP, the drug penetration process can be monitored to enable the image-guided cancer therapy. Both in vitro and in vivo assays prove the superior therapeutic efficacy of collagenase-enhanced photothermal and chemotherapy. The designed nanocomposite hydrogel therefore provides a versatile drug delivery system for deep tumor synergistic therapies.
Collapse
Affiliation(s)
- Yuru Chen
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China
| | - Ziteng Fan
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China
| | - Wenya Xu
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China
| | - Ziyi Zhu
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China
| | - Zhen Tan
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China
| | - Yaqin Hu
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China
| | - Irina Kurzina
- Laboratory of Translational Cellular and Molecular Biomedicine, Department of Natural Compounds, Pharmaceutical and Medicinal Chemistry, Department of Chemistry, National Research Tomsk State University, Tomsk 634050 Russia
| | - Nadezhda Cherdyntseva
- Laboratory of Translational Cellular and Molecular Biomedicine, Department of Natural Compounds, Pharmaceutical and Medicinal Chemistry, Department of Chemistry, National Research Tomsk State University, Tomsk 634050 Russia
| | - Wen Jing Yang
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China.
| | - Lianhui Wang
- Key Laboratory for Organic Electronics and Information Displays (KLOEID), Jiangsu Key Laboratory of Smart Biomaterials and Theranostic Technology, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023 China.
| |
Collapse
|
2
|
Pontoriero A, Critelli P, Zeppieri M, Bosurgi A, Guercio S, Caffo M, Angileri FF, Parisi S, Lavalle S, Pergolizzi S. Nano-drug delivery systems integrated with low radiation doses for enhanced therapeutic efficacy in cancer treatment. World J Clin Cases 2025; 13:101719. [DOI: 10.12998/wjcc.v13.i10.101719] [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: 09/24/2024] [Revised: 11/11/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Precision medicine is an emerging field that includes tumor-targeted delivery and tumor microenvironment. This review explores the synergistic potential of combining nano-drug delivery systems with low radiation doses to achieve optimized therapeutic outcomes, particularly in the context of cancer treatment. Nanoparticle-based drug carriers offer precise and targeted delivery, enhancing the therapeutic index of anticancer agents. The use of lower radiation doses has become a focus in radiation oncology to minimize off-target effects on healthy tissues in palliation treatment with high-target volume lesions.
AIM To conduct a bibliometric review of nanomedicine and glioblastoma (GBM), all relevant studies from the last two decades were included.
METHODS The search strategy comprised the keywords ”nanomedicine “and “glioblastoma” in the title and/or abstract. All English-language documents from 1 January 2000 to 31 December 2023 were considered for the analysis. R code (version 4.2.0) with R Studio (version 2022.12.0-353) and the Bibliometrix package (version 4.0.1) were used for the analysis. A total of 680 documents were collected.
RESULTS We analyzed the bibliometric features of nanomedicine in glioma. With the limitations of the research, our analysis aims to highlight the increasing interest of researchers in the precision medicine field in GBM treatment and lead us to suggest further studies focusing on the association between nanomedicine and radiotherapy.
CONCLUSION Due to the poor prognosis associated with GBM, new therapeutic approaches are necessary. There is an increasing interest in precision medicine, which includes nanomedicine and radiotherapy, for GBM treatment. This integration enhances the efficacy of targeted treatments and provides a promising avenue for reducing adverse effects, signifying a notable advancement in precision oncology.
Collapse
Affiliation(s)
- Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Paola Critelli
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, Udine 33100, Italy
| | - Alberto Bosurgi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Stefania Guercio
- Neurosurgery Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, Messina, Italy, Messina 98125, Italy
| | - Maria Caffo
- Neurosurgery Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, Messina, Italy, Messina 98125, Italy
| | - Filippo Flavio Angileri
- Neurosurgery Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, Messina, Italy, Messina 98125, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| | - Salvatore Lavalle
- Department of Medicine and Surgery, University of Enna "Kore", Enna 94100, Italy
| | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina 98125, Italy
| |
Collapse
|
3
|
Han X, Zhang X, Kang L, Feng S, Li Y, Zhao G. Peptide-modified nanoparticles for doxorubicin delivery: Strategies to overcome chemoresistance and perspectives on carbohydrate polymers. Int J Biol Macromol 2025; 299:140143. [PMID: 39855525 DOI: 10.1016/j.ijbiomac.2025.140143] [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: 12/16/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Chemotherapy serves as the primary treatment for cancers, facing challenges due to the emergence of drug resistance. Combination therapy has been developed to combat cancer drug resistance, yet it still suffers from lack of specific targeting of cancer cells and poor accumulation at the tumor site. Consequently, targeted administration of chemotherapy medications has been employed in cancer treatment. Doxorubicin (DOX) is one of the most frequently used chemotherapeutics, functioning by inhibiting topoisomerase activity. Enhancing the anti-cancer effects of DOX and overcoming drug resistance can be accomplished via delivery by nanoparticles. This review will focus on the development of peptide-DOX conjugates, the functionalization of nanoparticles with peptides, the co-delivery of DOX and peptides, as well as the theranostic use of peptide-modified nanoparticles in cancer treatment. The peptide-DOX conjugates have been designed to enhance the targeted delivery to cancer cells by interacting with receptors that are overexpressed on tumor surfaces. Moreover, nanoparticles can be modified with peptides to improve their uptake in tumor cells via endocytosis. Nanoparticles have the ability to co-deliver DOX along with therapeutic peptides for enhanced cancer treatment. Finally, nanoparticles modified with peptides can offer theranostic capabilities by facilitating both imaging and the delivery of DOX (chemotherapy).
Collapse
Affiliation(s)
- Xu Han
- Department of Traditional Chinese medicine, The First Hospital of China Medical University, Shenyang, China
| | - Xue Zhang
- Department of Gynecology, The First Hospital of China Medical University, Shenyang, China
| | - Longdan Kang
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Shuai Feng
- Department of Otolaryngology, The First Hospital of China Medical University, Shenyang, China.
| | - Yinyan Li
- Department of Ultrasonic Diagnosis, The First Hospital of China Medical University, Shenyang, China.
| | - Ge Zhao
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Kang H, Thomas RG, Kim S, Ju JK, Jeong YY. Anticancer therapeutic effect of magnetic guided cobalt ferrite/doxorubicin-loaded ROS-responsive bilirubin nanoparticles in a colon cancer model. Colloids Surf B Biointerfaces 2025; 248:114487. [PMID: 39756160 DOI: 10.1016/j.colsurfb.2024.114487] [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: 12/29/2023] [Revised: 11/19/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
PURPOSE The aim of this study is to synthesize the cobalt iron oxide (CoFe) and doxorubicin (Dox)-loaded chitosan bilirubin (ChiBil) nanoparticles and to investigate the anticancer therapeutic effect of the synthesized nanoparticles under magnetic guidance in a colon cancer. MATERIALS AND METHODS ChiBil-CoFe-Dox nanoparticles were synthesized by conjugating CoFe and Dox and then loaded onto ChiBil nanoparticles. Synthesis were characterized using thermogravimetric (TGA) analysis, inductive coupled plasma (ICP) analysis, dynamic light scattering (DLS), zeta potential and field emission-transmission electron microscopy (FE-TEM). Cellular uptake and cytotoxicity studies were conducted in vitro. Biodistribution and tumor inhibition study was done in vivo CT-26 colon cancer model. RESULTS The ChiBil-CoFe-Dox nanoparticles were successfully synthesized in this study. The in vitro cytotoxicity study showed that the ChiBil-CoFe-Dox nanoparticle had a toxic effect on cancer cells. The accumulation of ChiBil-CoFe-Dox nanoparticles was enhanced under magnetic guidance, as observed by in vivo. Tumor inhibition study showed that the ChiBil-CoFe-Dox nanoparticle effectively reduced tumor size in vivo mice colon cancer model, especially when combined with magnetic guidance. CONCLUSION This study showed that ChiBil-CoFe-Dox nanoparticle was successfully synthesized and effectively reduced tumor size, especially when combined with magnetic guidance. The in vitro and in vivo results suggested that the ROS stimuli responsive ChiBil-CoFe-Dox nanoparticles may be a potent therapeutic option for treating colon cancer.
Collapse
Affiliation(s)
- Hyo Kang
- Department of Surgery, Chonnam National University Medical School, 42, Jebong-ro, Dong-gu, Gwangju 61469, South Korea
| | - Reju George Thomas
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, South Korea.
| | - Subin Kim
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, South Korea
| | - Jae Kyun Ju
- Department of Surgery, Chonnam National University Medical School, 42, Jebong-ro, Dong-gu, Gwangju 61469, South Korea.
| | - Yong Yeon Jeong
- Department of Radiology, Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Hwasun, South Korea.
| |
Collapse
|
5
|
Menachem R, Nudelman I, Vorontsova A, Livneh I, Sela M, Benguigui M, Manobla B, Shammai Y, Deo A, Buxbaum C, Bessler R, Raviv Z, Shklover J, Sznitman J, Ciechanover A, Schroeder A, Shaked Y. Bone Marrow-Targeted Liposomes Loaded with Bortezomib Overcome Multiple Myeloma Resistance. ACS NANO 2025. [PMID: 40117329 DOI: 10.1021/acsnano.4c10597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Multiple myeloma (MM) poses a significant therapeutic challenge due to its persistent progression and low survival rate. Although the proteasome inhibitor bortezomib has revolutionized MM treatment, MM aggressiveness and drug resistance remain critical concerns. To tackle this problem, we developed AMD3100-targeted Bortezomib Liposomes (ATBL) designed for the targeted delivery of bortezomib to MM cells. Uptake of ATBL into MM cells was dependent on CXCR4 and was enhanced compared to nontargeted liposomes, both in vitro and in vivo. Treating MM-bearing mice with ATBL achieved superior therapeutic efficacy compared to treatment with free bortezomib or nontargeted bortezomib-loaded liposomes. Notably, the therapeutic activity of ATBL was limited in mice inoculated with CXCR4-knockdown MM cells, highlighting CXCR4 as a potential biomarker for ATBL response. Importantly, ATBL was effective against an aggressive and bortezomib-resistant MM clone both in vitro and in vivo. Toxicity and biodistribution profiles demonstrated the safety and bone marrow-targeting ability of ATBL. Collectively, this study highlights ATBL as a promising next-generation proteasome inhibitor-based therapy that incorporates bone marrow-targeting ability and sensitizing elements to overcome drug resistance in MM.
Collapse
Affiliation(s)
- Rotem Menachem
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- Faculty of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Igor Nudelman
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- Faculty of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Avital Vorontsova
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Ido Livneh
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Mor Sela
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- Faculty of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Madeleine Benguigui
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Bar Manobla
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Yael Shammai
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- Faculty of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Abhilash Deo
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Chen Buxbaum
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Ron Bessler
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200001, Israel
| | - Ziv Raviv
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Jeny Shklover
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- Faculty of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Josué Sznitman
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa 3200001, Israel
| | - Aaron Ciechanover
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| | - Avi Schroeder
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
- Faculty of Chemical Engineering, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Yuval Shaked
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3525422, Israel
- Rappaport Technion Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa 3525422, Israel
| |
Collapse
|
6
|
Fang L, Zhu R, Li M, Ma J, Fan S, He X, Yang Z, Yan Y, Ma X, Xiang G. Silica-based EGFR-degrading nano-PROTACs for efficient therapy of non-small cell lung cancer. Eur J Pharm Biopharm 2025:114699. [PMID: 40113047 DOI: 10.1016/j.ejpb.2025.114699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/22/2025]
Abstract
Proteolysis targeting chimeras (PROTACs) technology is a promising strategy for degrading proteins of interest. Traditional PROTACs, however, often face challenges such as poor solubility, low stability, and off-target toxicity. To address these challenges, we integrated nanotechnology to enhance the delivery of target-protein degraders to the tumor sites, thereby improving their properties. Here, we report silica-based nano-PROTACs (SiPROTACs) that feature multiple ligands on the surface to target and degrade the transmembrane protein epidermal growth factor receptor (EGFR). SiPROTACs, with a diameter of approximately 50 nm, can efficiently bind to EGFR, recruit cereblon (CRBN) to induce EGFR ubiquitination, and facilitate their degradation by proteasomes. In HCC-827 and PC-9 cell lines, SiPROTACs initiated EGFR degradation at a notably low concentration of 50 nM, demonstrating greater efficiency compared to traditional PROTACs. In HCC-827 xenograft tumor-bearing mice, SiPROTACs accumulated at tumor site for at least 48 h and exhibited significant anti-tumor effects in vivo without causing noticeable side effects. These findings suggest a novel approach for the application of PROTACs highlighting their therapeutic potential for the treatment of non-small cell lung cancer (NSCLC).
Collapse
Affiliation(s)
- Lei Fang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ruixue Zhu
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meijing Li
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Junhui Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sijun Fan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xuelian He
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhongrui Yang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yakai Yan
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiang Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Departement of Pharmacy, Tongren Polytechnic College, Tongren 554300, China.
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Departement of Pharmacy, Tongren Polytechnic College, Tongren 554300, China; Institute of the Higher Education Edible and Medicinal Fungi Engineering Research Center, Tongren 554300, China.
| |
Collapse
|
7
|
Li Y, Liu P, Zhang B, Chen J, Yan Y. Global trends and research hotspots in nanodrug delivery systems for breast cancer therapy: a bibliometric analysis (2013-2023). Discov Oncol 2025; 16:269. [PMID: 40047951 PMCID: PMC11885776 DOI: 10.1007/s12672-025-02014-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
OBJECTIVE Nanomedicine offers fresh approaches for breast cancer treatment, countering traditional limitations. The nanodrug delivery system's precision and biocompatibility hold promise, yet integration hurdles remain. This study reviews nano delivery systems in breast cancer therapy from 2013 to 2023, guiding future research directions. METHODS In this study, we conducted a comprehensive search on Web of Science database (Guilin Medical University purchase edition) and downloaded literature related to the field published between 2013 and 2023. We analyzed these publications using R software, VOSviewer, and CiteSpace software. RESULTS This study reviewed 2632 documents, showing a steady publication increase from 2013 to 2023, peaking at 408 in 2022. China, USA, India, and Iran were prominent in publishing. The Chinese Academy of Sciences and Tabriz University of Medical Science were key collaboration centers. Notably, the Journal of Controlled Release and Biomaterials ranked among the top 10 journals for publications and citations, establishing their field representation. Key terms like "breast cancer," "nanoparticles," "drug delivery," "in-vitro," and "delivery" were widely used. Research focused on optimizing drug targeting, utilizing the tumor microenvironment for drug delivery, and improving delivery efficiency. CONCLUSION The nanodrug delivery system, as an innovative drug delivery approach, offers numerous advantages and has garnered global attention from researchers. This study provides an analysis of the status and hotspots in nano delivery systems within the realm of breast cancer therapy, offering valuable insights for future research in this domain.
Collapse
Affiliation(s)
- Yang Li
- Department of Pharmacy, The First People's Hospital of Yulin, Yulin, Guangxi, China
| | - Pingping Liu
- Sanya Central Hospital (The Third People's Hospital of Hainan Province), Hainan, China
| | - Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Juan Chen
- Sanya Central Hospital (The Third People's Hospital of Hainan Province), Hainan, China
| | - Yuanyuan Yan
- Sanya Central Hospital (The Third People's Hospital of Hainan Province), Hainan, China.
| |
Collapse
|
8
|
Batool A, Kopp I, Kubeil M, Bachmann M, Andrews PC, Stephan H. Targeted bismuth-based materials for cancer. Dalton Trans 2025. [PMID: 40040450 DOI: 10.1039/d5dt00163c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
The use of bismuth and its compounds in biomedicine has developed rapidly in recent years. Due to their unique properties, there are great opportunities for the development of new non-invasive strategies for the early diagnosis and effective treatment of cancers. This perspective highlights key fabrication methods to generate well-defined and clinically relevant bismuth materials of varying characteristics. On the one hand, this opens up a wide range of possibilities for unimodal and multimodal imaging. On the other hand, effective treatment strategies, which are increasingly based on combinatorial therapies, are given a great deal of attention. One of the biggest challenges remains the selective tumour targeting, whether active or passive. Here we present an overview on new developments of bismuth based materials moving forward from a simple enrichment at the tumour site via uptake by the mononuclear phagocytic system (MPS) to a more active tumour specific targeting via covalent modification with tumour-seeking molecules based on either small or antibody-derived molecules.
Collapse
Affiliation(s)
- Amna Batool
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Ina Kopp
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Manja Kubeil
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Philip C Andrews
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
| | - Holger Stephan
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| |
Collapse
|
9
|
Hussein L, Moaness M, Mabrouk M, Farahat MG, Beherei HH. Advancements in mesoporous bioactive glasses for effective bone cancer therapy: Recent developments and future perspectives. BIOMATERIALS AND BIOSYSTEMS 2025; 17:100108. [PMID: 40083816 PMCID: PMC11904600 DOI: 10.1016/j.bbiosy.2025.100108] [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: 12/30/2024] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
This review focuses on recent advancements in the effective use of mesoporous bioactive glasses (MBG) in the treatment of bone cancer, focusing on Osteosarcoma (OS). Bone cancers are rare but are associated with significant morbidity and mortality; often, aggressive treatment is required. Conventional treatments such as surgery, radiation, and chemotherapy are often not enough. This is because surgery cannot completely remove the tumor, without creating a critical size which are defects larger than 2 cm that cannot be repaired by physiological mechanisms. As a result, patients often face the additional burden of radiation and chemotherapy. Scientists have been exploring new treatments, including hyperthermia-targeted therapy, polymeric nanoparticles, and stem cell therapy. This could potentially negatively impact healthy tissues and organs. MBG offers a promising alternative to chemotherapeutic agents and ions for disease treatment as it acts as a multifunctional drug delivery system (DDS). In addition, MBG can also be engineered into scaffolds to facilitate local delivery of growth factors and drugs, thus promoting the efficiency of bone healing and restoration. Therefore, the current review highlights various MBG types reported in the past decade and explores potential future paths to enhance their use in bone cancer treatment while also giving insight on the already commercially available BGs that are used in different bone-related disease.
Collapse
Affiliation(s)
- Laila Hussein
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mona Moaness
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mohamed G. Farahat
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza, Egypt
- Biotechnology Department, Faculty of Postgraduate Studies for Nanotechnology, Sheikh Zayed Branch Campus, Cairo University, Sheikh Zayed City 12588, Egypt
| | - Hanan H. Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| |
Collapse
|
10
|
Yang H, Li J, Song C, Li H, Luo Q, Chen M. Emerging Gene Therapy Based on Nanocarriers: A Promising Therapeutic Alternative for Cardiovascular Diseases and a Novel Strategy in Valvular Heart Disease. Int J Mol Sci 2025; 26:1743. [PMID: 40004206 PMCID: PMC11855571 DOI: 10.3390/ijms26041743] [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: 12/31/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular disease remains a leading cause of global mortality, with many unresolved issues in current clinical treatment strategies despite years of extensive research. Due to the great progress in nanotechnology and gene therapy in recent years, the emerging gene therapy based on nanocarriers has provided a promising therapeutic alternative for cardiovascular diseases. This review outlines the status of nanocarriers as vectors in gene therapy for cardiovascular diseases, including coronary heart disease, pulmonary hypertension, hypertension, and valvular heart disease. It discusses challenges and future prospects, aiming to support emerging clinical treatments. This review is the first to summarize gene therapy using nanocarriers for valvular heart disease, highlighting their potential in targeting challenging tissues.
Collapse
Affiliation(s)
- Haoran Yang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chengxiang Song
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Hongde Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
| | - Qiang Luo
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; (H.Y.); (J.L.); (C.S.)
- Department of Cardiology, West China Hospital, Sichuan University, No.37 Guoxue Street, Chengdu 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Garg S, Singla P, Kaur S, Canfarotta F, Velliou E, Dawson JA, Kapur N, Warren NJ, Amarnath S, Peeters M. Future Perspectives on the Automation and Biocompatibility of Molecularly Imprinted Polymers for Healthcare Applications. Macromolecules 2025; 58:1157-1168. [PMID: 39958488 PMCID: PMC11823616 DOI: 10.1021/acs.macromol.4c01621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 02/18/2025]
Abstract
Molecular recognition is of crucial importance in several healthcare applications, such as sensing, drug delivery, and therapeutics. Molecularly imprinted polymers (MIPs) present an interesting alternative to biological receptors (e.g., antibodies, enzymes) for this purpose since synthetic receptors overcome the limited robustness, flexibility, high-cost, and potential for inhibition that comes with natural recognition elements. However, off the shelf MIP products remain limited, which is likely due to the lack of a scalable production approach that can manufacture these materials in high yields and narrow and defined size distributions to have full control over their properties. In this Perspective, we will confer how breakthroughs in the automation of MIP design, manufacturing, and evaluation of performance will accelerate the (commercial) implementation of MIPs in healthcare technology. In addition, we will discuss how prediction of the in vivo behavior of MIPs with animal-free technologies (e.g., 3D tissue models) will be critical to assess their clinical potential.
Collapse
Affiliation(s)
- Saweta Garg
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Pankaj Singla
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
| | - Sarbjeet Kaur
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Francesco Canfarotta
- MIP
Discovery, Colworth Park, Sharnbrook, MK44 1LQ, Bedfordshire, United Kingdom
| | - Eirini Velliou
- University
College London, Centre for 3D Models
of Health and Disease, Charles Bell House, London, W1W 7TY, United Kingdom
| | - James A. Dawson
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Nikil Kapur
- University
of Leeds, School of Mechanical Engineering, Woodhouse Lane, Leeds, LS2 9JT, United Kingdom
| | - Nicholas J. Warren
- School of
Chemical, Materials and Biological Engineering, University of Sheffield, Sir Robert Hadfield Building, Mappin Street, Sheffield, S1 3JD, United Kingdom
| | - Shoba Amarnath
- Newcastle
University, Newcastle
upon Tyne, Tyne and Wear, NE1 7RU, United Kingdom
| | - Marloes Peeters
- University
of Manchester, School of Engineering, Engineering A Building, Booth East
Street, Manchester, M13
9QS, United Kingdom
| |
Collapse
|
12
|
Dijkstra M, Schueffl H, Adamova B, Baumfried O, Kastner A, Berger W, Keppler BK, Heffeter P, Kowol CR. Exploring the Structure-Activity Relationships of Albumin-Targeted Picoplatin-Based Platinum(IV) Prodrugs. Inorg Chem 2025; 64:2554-2566. [PMID: 39878587 DOI: 10.1021/acs.inorgchem.4c05269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Platinum(II) complexes prevail as first-line treatment for many cancers but are associated with serious side effects and resistance development. Picoplatin emerged as a promising alternative to circumvent GSH-induced tumor resistance by introducing a bulky 2-picoline ligand. Although clinical studies were encouraging, picoplatin did not receive approval. Interestingly, the anticancer potential of prodrugs based on picoplatin is widely underexplored, and even less so the respective tumor-targeting approaches. We synthesized two new "hybrid" picoplatin(II) derivatives with an oxalate or cyclobutane dicarboxylate leaving group and their corresponding platinum(IV) prodrugs with an albumin-targeting maleimide moiety or a succinimide as reference. Picoplatin(II) and its derivatives indeed reacted much slower with GSH compared to the respective analogs cisplatin, carboplatin, or oxaliplatin. While PicoCarbo(IV) and PicoOxali(IV) were reduced slowly in the presence of ascorbic acid, picoplatin(IV) was extremely unstable. All three prodrugs were widely inactive in the MTT assays. The platinum(IV)-maleimide complexes rapidly bound to albumin with stable conjugates for >25 h. Albumin-binding resulted in elevated platinum plasma levels, prolonged blood circulation, and enhanced tumor accumulation of the prodrugs in mice bearing CT26 tumors. However, only maleimide-functionalized PicoCarbo(IV) and picoplatin(II) significantly inhibited tumor growth. One possible explanation is that for albumin-binding platinum(IV) prodrugs, the bulky 2-picoline moiety prevents sufficient activation/reduction to unlock their full anticancer potential.
Collapse
Affiliation(s)
- Martijn Dijkstra
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria
| | - Hemma Schueffl
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Barbora Adamova
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Oliver Baumfried
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
| | - Alexander Kastner
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", 1090 Vienna, Austria
| | - Bernhard K Keppler
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", 1090 Vienna, Austria
| | - Petra Heffeter
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090 Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", 1090 Vienna, Austria
| | - Christian R Kowol
- Faculty of Chemistry, Institute of Inorganic Chemistry, University of Vienna, Waehringer Str. 42, 1090 Vienna, Austria
- Research Cluster "Translational Cancer Therapy Research", 1090 Vienna, Austria
| |
Collapse
|
13
|
Malinovskaya J, Kovshova T, Melnikov P, Li Z, Dhakal N, Knoll J, Valikhov M, Ermolenko Y, Chernysheva A, Gurina O, Chekhonin V, Wacker MG, Gelperina S. The second phase of tumor invasion driven by immune cells: A study on doxorubicin-loaded PLG nanoparticles. J Control Release 2025; 378:750-762. [PMID: 39724952 DOI: 10.1016/j.jconrel.2024.12.056] [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: 10/22/2024] [Revised: 12/01/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
Poly(lactide-co-glycolide) (PLG) nanoparticles loaded with doxorubicin have reached phase-I clinical trials for treating advanced solid tumors. This study explores cell hitchhiking, where nanoparticles associate with blood cells and investigates the impact on pharmacokinetics and tumor migration. Previous findings highlighted the early post-injection phase dominated by nonspecific nanoparticle-cell interactions and burst release. In contrast, this study examines the subsequent phase of tumor invasion, emphasizing the role of immune cells, mostly neutrophils, in redistributing the carrier to the tumor site via blood cell hitchhiking. We provide a detailed investigation of nanoparticle extravasation kinetics and mechanisms, showing qualitative and quantitative evidence of increased nanoparticle association with immune cells over time. By 30 min post-injection, approximately 15 % of monocytes and 15-19 % of neutrophils tested positive for nanoparticles, with significant differences observed between ex vivo and in vivo experiments, and between healthy and tumor-bearing animals. This study underscores the ambiguous role of immune cell-mediated tumor targeting. While the total accumulation of the carrier rises, this fraction is partially trapped in immune cells without any chance to escape.
Collapse
Affiliation(s)
- Julia Malinovskaya
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia
| | - Tatyana Kovshova
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia
| | - Pavel Melnikov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Zhuoxuan Li
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore
| | - Namrata Dhakal
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore
| | - Julian Knoll
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore
| | - Marat Valikhov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Yulia Ermolenko
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia
| | - Anastasia Chernysheva
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Olga Gurina
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Matthias G Wacker
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore..
| | - Svetlana Gelperina
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia.
| |
Collapse
|
14
|
Tang L, Yang X, He L, Zhu C, Chen Q. Preclinical advance in nanoliposome-mediated photothermal therapy in liver cancer. Lipids Health Dis 2025; 24:31. [PMID: 39891269 PMCID: PMC11783920 DOI: 10.1186/s12944-024-02429-x] [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: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/03/2025] Open
Abstract
Liver cancer is a highly lethal malignant tumor with a high incidence worldwide. Therefore, its treatment has long been a focus of medical research. Although traditional treatment methods such as surgery, radiotherapy, and chemotherapy have increased the survival rate of patients, their efficacy remains unsatisfactory owing to the nonspecific distribution of drugs, high toxicity, and drug resistance of tumor tissues. In recent years, the application of nanotechnology in the medical field has opened a new avenue for the treatment of liver cancer. Among these treatment methods, photothermal therapy (PTT) based on nanoliposomes has attracted wide attention owing to its unique targeting and high efficiency. This article reviews the latest preclinical research progress of nanoliposome-based PTT for liver cancer and its metastasis, discusses the preclinical challenges in this field, and proposes directions for improvement, with the aim of improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Lixuan Tang
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiao Yang
- The department of oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liwen He
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chaogeng Zhu
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Qingshan Chen
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
15
|
Izadiyan Z, Misran M, Kalantari K, Webster TJ, Kia P, Basrowi NA, Rasouli E, Shameli K. Advancements in Liposomal Nanomedicines: Innovative Formulations, Therapeutic Applications, and Future Directions in Precision Medicine. Int J Nanomedicine 2025; 20:1213-1262. [PMID: 39911259 PMCID: PMC11794392 DOI: 10.2147/ijn.s488961] [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: 07/27/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Liposomal nanomedicines have emerged as a pivotal approach for the treatment of various diseases, notably cancer and infectious diseases. This manuscript provides an in-depth review of recent advancements in liposomal formulations, highlighting their composition, targeted delivery strategies, and mechanisms of action. We explore the evolution of liposomal products currently in clinical trials, emphasizing their potential in addressing diverse medical challenges. The integration of immunotherapeutic agents within liposomes marks a paradigm shift, enabling the design of 'immuno-modulatory hubs' capable of orchestrating precise immune responses while facilitating theranostic applications. The recent COVID-19 pandemic has accelerated research in liposomal-based vaccines and antiviral therapies, underscoring the need for improved delivery mechanisms to overcome challenges like rapid clearance and organ toxicity. Furthermore, we discuss the potential of "smart" liposomes, which can respond to specific disease microenvironments, enhancing treatment efficacy and precision. The integration of artificial intelligence and machine learning in optimizing liposomal designs promises to revolutionize personalized medicine, paving the way for innovative strategies in disease detection and therapeutic interventions. This comprehensive review underscores the significance of ongoing research in liposomal technologies, with implications for future clinical applications and enhanced patient outcomes.
Collapse
Affiliation(s)
- Zahra Izadiyan
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Misni Misran
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Katayoon Kalantari
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Thomas J Webster
- Biomedical Engineering, Hebei University of Technology, Tianjin, People’s Republic of China
- School of Engineering, Saveetha University, Chennai, India
| | - Pooneh Kia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Elisa Rasouli
- Department of Electrical and Electronics Engineering, Nanyang Technological University, Nanyang, Singapore
| | - Kamyar Shameli
- School of Medicine, Institute of Virology, Technical University of Munich, Munich, Germany
| |
Collapse
|
16
|
Kojima C, Hirata R, Dei N, He H, Ikemoto Y, Matsumoto A. Hydration and Biodistribution of Zwitterionic Dendrimers Conjugating a Sulfobetaine Monomer and Polymers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:1411-1417. [PMID: 39778908 PMCID: PMC11755784 DOI: 10.1021/acs.langmuir.4c04276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
Zwitterionic polymers exhibit strong hydration, high biocompatibility, and antifouling properties. Dendrimers are regularly branched polymers, which are used in the drug delivery system (DDS). In this study, we synthesized zwitterionic monomer- and polymer-conjugated dendrimers as a biocompatible nanoparticle to investigate the relation between the hydration property and biodistribution. A sulfobetaine monomer (SBM) was conjugated at the termini of the polyamidoamine (PAMAM) dendrimer. Polysulfobetaines (PSBs) were produced by reversible addition-fragmentation chain transfer polymerization and were also conjugated at the termini. Intermediate water, that is, water molecules loosely bound to the material, can be estimated from the melting peaks at less than 0 °C in differential scanning calorimetry (DSC) measurement. Our DSC results showed that the PSB-conjugated dendrimers (PSM-dens) contained more intermediate water than the SBM-conjugated dendrimer (SBM-den). PSB-dens accumulated in the tumor after intravenous administration, but SBM-den did not. These suggested that the amount of intermediate water, that is, the hydration property, was related to the biodistribution of the zwitterionic dendrimers. This relation is a possible design criterion for drug carriers. PSB-dens accumulated in the tumor even after the second injection, possibly overcoming the accelerated blood clearance observed with poly(ethylene glycol)-modified nanoparticles. Thus, this kind of zwitterionic polymer-conjugated dendrimer is useful for the DDS in cancer treatment.
Collapse
Affiliation(s)
- Chie Kojima
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Rikuto Hirata
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Nanako Dei
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| | - Hao He
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
- Department of Materials Science and Engineering, School of Materials and Chemical Technology, Institute of Science Tokyo, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Yuka Ikemoto
- Spectroscopy Division, Japan Synchrotron Radiation Research Institute, 1-1-1 Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5198, Japan
| | - Akikazu Matsumoto
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Metropolitan University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 599-8531, Japan
| |
Collapse
|
17
|
Razavi R, Khajouei G, Divsalar F, Dawi E, Amiri M. Recent advances on brain drug delivery via nanoparticles: alternative future materials for neuroscience applications; a review. Rev Neurosci 2025:revneuro-2024-0086. [PMID: 39829237 DOI: 10.1515/revneuro-2024-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/29/2024] [Indexed: 01/22/2025]
Abstract
Essentially, the blood-brain barrier (BBB) serves as a line of demarcation between neural tissues and the bloodstream. A unique and protective characteristic of the blood-brain barrier is its ability to maintain cerebral homeostasis by regulating the flux of molecules and ions. The inability to uphold proper functioning in any of these constituents leads to the disruption of this specialized multicellular arrangement, consequently fostering neuroinflammation and neurodegeneration. Recent advancements in nanomedicine have been regarded as a promising avenue for improving the delivery of drugs to the central nervous system in the modern era. A major benefit of this innovation is that it allows drugs to accumulate selectively within the cerebral area by circumventing the blood-brain barrier. Although brain-targeted nanomedicines have demonstrated impressive achievements, certain limitations in targeting specificity still exist. In this examination, we scrutinize the distinctive physical and chemical attributes of nanoparticles (NPs) contributing to their facilitation in BBB traversal. We explore the various mechanisms governing NP passage over the BBB, encompassing paracellular conveyance, mediated transport, as well as adsorptive- and receptor-mediated transcytosis. The therapeutic success of NPs for the treatment of brain tumors has been extensively investigated through the use of various categories of NPs. Among these are polymeric nanoparticles, liposomes, solid lipid nanoparticles, dendrimers, metallic nanoparticles, quantum dots, and nanogels. The potential utility of nanoparticles goes beyond their ability to transport pharmaceuticals. They can serve as adept imaging contrast agents, capable of being linked with imaging probes. This will facilitate tumor visualization, delineate lesion boundaries and margins, and monitor drug delivery and treatment response. Versatile nanoparticles can be engineered to effectively target neoplastic lesions, serving dual roles in diagnostic imaging and therapeutic interventions. Subsequently, this discourse explores the constraints associated with nanoparticles in the context of treating brain tumors.
Collapse
Affiliation(s)
- Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran
| | - Ghazal Khajouei
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | - Fatemeh Divsalar
- Sina Hospital, Zarand Network and Health Center, 48463 Kerman University of Medical Sciences , Kerman, Iran
| | - Elmuez Dawi
- College of Humanities and Sciences, College of Humanities and Sciences, Department of Mathematics and Sciences, Ajman University, P.O. Box 346, Ajman, United Arab Emirates
| | - Mahnaz Amiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| |
Collapse
|
18
|
Meng JL, Dong ZX, Chen YR, Lin MH, Liu YC, Roffler SR, Lin WW, Chang CY, Tzou SC, Cheng TL, Huang HC, Li ZQ, Lin YC, Su YC. pH-Responsive Polyethylene Glycol Engagers for Enhanced Brain Delivery of PEGylated Nanomedicine to Treat Glioblastoma. ACS NANO 2025; 19:307-321. [PMID: 39749925 PMCID: PMC11752499 DOI: 10.1021/acsnano.4c05906] [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: 05/04/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
The blood-brain barrier (BBB) remains a major obstacle for effective delivery of therapeutics to treat central nervous system (CNS) disorders. Although transferrin receptor (TfR)-mediated transcytosis is widely employed for brain drug delivery, the inefficient release of therapeutic payload hinders their efficacy from crossing the BBB. Here, we developed a pH-responsive anti-polyethylene glycol (PEG) × anti-TfR bispecific antibody (pH-PEG engagerTfR) that can complex with PEGylated nanomedicine at physiological pH to trigger TfR-mediated transcytosis in the brain microvascular endothelial cells, while rapidly dissociating from PEGylated nanomedicine at acidic endosomes for efficient release of PEGylated nanomedicine to cross the BBB. The pH-PEG engagerTfR significantly increased the accumulation of PEGylated nanomedicine in the mouse brain compared to wild-type PEG engagerTfR (WT-PEG engagerTfR). pH-PEG engagerTfR-decorated PEGylated liposomal doxorubicin exhibited an enhanced antitumor effect and extended survival in a human glioblastoma (GBM) orthotopic xenograft mice model. Conditional release of PEGylated nanomedicine during BBB-related receptor-mediated transcytosis by pH-PEG engagerTfR is promising for enhanced brain drug delivery to treat CNS disorders.
Collapse
Affiliation(s)
- Jun-Lun Meng
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zi-Xuan Dong
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yan-Ru Chen
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Meng-Hsuan Lin
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Ching Liu
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Steve R. Roffler
- Institute
of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
- Graduate
Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wen-Wei Lin
- School
of Post-Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chin-Yuan Chang
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Shey-Cherng Tzou
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department
of Biomedical Science and Environmental Biology, Drug Development
and Value Creation Research Center, Kaohsiung
Medical University, Kaohsiung 807, Taiwan
| | - Tian-Lu Cheng
- Department
of Biomedical Science and Environmental Biology, Drug Development
and Value Creation Research Center, Kaohsiung
Medical University, Kaohsiung 807, Taiwan
| | - Hsiao-Chen Huang
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Zhi-Qin Li
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yen-Cheng Lin
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Cheng Su
- Department
of Biological Science and Technology, Center for Intelligent Drug
Systems and Smart Bio-devices (IDSB), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department
of Biomedical Science and Environmental Biology, Drug Development
and Value Creation Research Center, Kaohsiung
Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
19
|
Lv MY, Hou DY, Liu SW, Cheng DB, Wang H. Strategy and Design of In Situ Activated Protein Hydrolysis Targeted Chimeras. ACS NANO 2025; 19:101-119. [PMID: 39731609 DOI: 10.1021/acsnano.4c11903] [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: 12/30/2024]
Abstract
Protein hydrolysis targeted chimeras (PROTACs) represent a different therapeutic approach, particularly relevant for overcoming challenges associated with traditional small molecule inhibitors. These challenges include targeting difficult proteins that are often deemed "undruggable" and addressing issues of acquired resistance. PROTACs employ the body's own E3 ubiquitin ligases to induce the degradation of specific proteins of interest (POIs) through the ubiquitin-proteasome pathway. This process is cyclical, allowing for broad applicability, potent protein degradation, and selective targeting. Despite their effectiveness, PROTACs can inadvertently target and degrade nonspecific proteins, potentially resulting in significant side effects and off-target toxicity. To address this concern, researchers have created stimuli-activated PROTACs that enhance targeted protein degradation while minimizing potential harm to healthy cells. These advanced PROTACs aim to improve the precision of degradation in both time and space. This article reviews the strategies for in situ activated PROTACs, highlighting key compounds and research advancements associated with various mechanisms of action. The insights presented here aim to guide further exploration in the field of activated PROTACs.
Collapse
Affiliation(s)
- Mei-Yu Lv
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Harbin 150001, China
| | - Da-Yong Hou
- Department of PET-CT/MRI, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Harbin 150001, China
| | - Shao-Wei Liu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, P. R. China
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, P. R. China
| | - Haoran Wang
- Faculty of Materials Science, Shenzhen MSU-BIT University, Shenzhen 518100, P. R. China
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon 999077, Hong Kong, China
| |
Collapse
|
20
|
Ding Q, Chen H, Zhang Y, Yang J, Li M, He Q, Mei L. Innovative integration of nanomedicines and phototherapy to modulate autophagy for enhanced tumor eradication. J Control Release 2025; 377:855-879. [PMID: 39631701 DOI: 10.1016/j.jconrel.2024.11.071] [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: 10/24/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Nanomedicines, by significantly enhancing the solubility, stability, and targeted delivery of therapeutic agents, have emerged as transformative tools in light-induced therapies, particularly in the context of oncology. These advancements are attributed to their ability to mediate autophagy through light activation, thereby revolutionizing cancer treatment paradigms. This review provides a comprehensive analysis of the state-of-the-art integration of nanomedicines with phototherapy techniques, emphasizing their role in modulating autophagy within cancer cells. It delineates the potential of light-responsive nanomaterials to induce selective tumor cell death by precisely regulating over-activated autophagy pathways. Additionally, it discusses innovative strategies for combining nanomedicines with phototherapy and other clinical modalities for tumor treatment, as well as integrating autophagy with various forms of programmed cell death to address challenges related to drug resistance and therapeutic efficacy. By synthesizing recent advancements and delineating future research directions, this review offers a thorough perspective on the optimization of light-induced autophagy through nanomedicines, highlighting novel strategies for enhancing cancer treatment efficacy.
Collapse
Affiliation(s)
- Qihang Ding
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China; Department of Chemistry, Korea University, Seoul 02841, South Korea
| | - Haiyan Chen
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Yifan Zhang
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Junbin Yang
- Hainan Academy of Inspection and Testing, Hainan 570203, PR China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, PR China.
| | - Ling Mei
- Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
21
|
Pérez-Pacheco Y, Tylkowski B, García-Valls R. Chitosan Micro/Nanocapsules in Action: Linking Design, Production, and Therapeutic Application. Molecules 2025; 30:252. [PMID: 39860124 PMCID: PMC11767700 DOI: 10.3390/molecules30020252] [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: 12/12/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
pH sensitivity of chitosan allows for precise phase transitions in acidic environments, controlling swelling and shrinking, making chitosan suitable for drug delivery systems. pH transitions are modulated by the presence of cross-linkers by the functionalization of the chitosan chain. This review relays a summary of chitosan functionalization and tailoring to optimize drug release. The potential to customize chitosan for different environments and therapeutic uses introduces opportunities for drug encapsulation and release. The focus on improving drug encapsulation and sustained release in specific tissues is an advanced interpretation, reflecting the evolving role of chitosan in achieving targeted and more efficient therapeutic outcomes. This review describes strategies to improve solubility and stability and ensure the controlled release of encapsulated drugs. The discussion on optimizing factors like cross-linking density, particle size, and pH for controlled drug release introduces a deeper understanding of how to achieve specific therapeutic effects. These strategies represent a refined approach to designing chitosan-based systems, pushing the boundaries of sustained release technologies and offering new avenues for precise drug delivery profiles.
Collapse
Affiliation(s)
- Yaride Pérez-Pacheco
- Department of Chemical Engineering, Universitat Rovira i Virgili, Av. Països Catalans 26, Campus Sescelades, 43007 Tarragona, Spain; (Y.P.-P.); (B.T.)
| | - Bartosz Tylkowski
- Department of Chemical Engineering, Universitat Rovira i Virgili, Av. Països Catalans 26, Campus Sescelades, 43007 Tarragona, Spain; (Y.P.-P.); (B.T.)
- Eurecat, Centre Tecnològic de Catalunya, Chemical Technologies Unit, Marcel_lí Domingo s/n, 43007 Tarragona, Spain
- Faculty of Health Science, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, ul. Sklodowskiej Curie 9, 85-094 Bydgoszcz, Poland
| | - Ricard García-Valls
- Department of Chemical Engineering, Universitat Rovira i Virgili, Av. Països Catalans 26, Campus Sescelades, 43007 Tarragona, Spain; (Y.P.-P.); (B.T.)
| |
Collapse
|
22
|
Spiegelberg D, Hwang LA, Pua KH, Kumar SC, Koh XY, Koh XH, Selvaraju RK, Sabapathy K, Nestor M, Lane D. Targeting mutant p53: Evaluation of novel anti-p53 R175H monoclonal antibodies as diagnostic tools. Sci Rep 2025; 15:1000. [PMID: 39762369 PMCID: PMC11704002 DOI: 10.1038/s41598-024-83871-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
About 50% of all cancers carry a mutation in p53 that impairs its tumor suppressor function. The p53 missense mutation p53R175H (p53R172H in mice) is a hotspot mutation in various cancer types. Therefore, monoclonal antibodies selectively targeting clinically relevant mutations like p53R175H could prove immensely value. We aimed to evaluate the in vitro and in vivo binding properties of two novel anti-p53R175H monoclonal antibodies and to assess their performance as agents for molecular imaging. In vitro, 125I-4H5 and 125I-7B9 demonstrated long shelf life and antigen-specific binding. Our in vivo study design allowed head-to-head comparison of the antibodies in a double tumor model using repeated SPECT/CT imaging, followed by biodistribution and autoradiography. Both tracers performed similarly, with marginally faster blood clearance for 125I-7B9. Repeated molecular imaging demonstrated suitable imaging characteristics for both antibodies, with the best contrast images occurring at 48 h post-injection. Significantly higher uptake was detected in the mut-p53-expressing tumors, confirmed by ex vivo autoradiography. We conclude that molecular imaging with an anti-p53R175H tracer could be a promising approach for cancer diagnostics and could be further applied for patient stratification and treatment response monitoring of mutant p53-targeted therapeutics.
Collapse
Affiliation(s)
- Diana Spiegelberg
- Department of Immunology, Genetics, Pathology, Uppsala University, Uppsala, Sweden.
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| | - Le-Ann Hwang
- Divisions of Cellular & Molecular Research, National Cancer Centre Singapore, Singapore, 168583, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Khian Hong Pua
- Institute of Molecular and Cellular Biology, ASTAR, Singapore, 138673, Singapore
| | - Sashwini Chandra Kumar
- Divisions of Cellular & Molecular Research, National Cancer Centre Singapore, Singapore, 168583, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Xin Yu Koh
- Institute of Molecular and Cellular Biology, ASTAR, Singapore, 138673, Singapore
| | - Xiao Hui Koh
- Institute of Molecular and Cellular Biology, ASTAR, Singapore, 138673, Singapore
| | - Ram Kumar Selvaraju
- Preclinical PET-MRI Platform, Part of Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Kanaga Sabapathy
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Marika Nestor
- Department of Immunology, Genetics, Pathology, Uppsala University, Uppsala, Sweden
| | - David Lane
- Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Rahaman W, Chaudhuri A. Self-assembled Lipid Nanoparticles for Killing Triple Negative Breast Cancer Cells. Chem Asian J 2025; 20:e202401049. [PMID: 39466002 DOI: 10.1002/asia.202401049] [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: 08/22/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Triple negative breast cancers (TNBCs) lacking estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) on their cell surfaces are highly aggressive, difficult-to-treat and often relapse. Herein, we report on the self-assembled lipid nanoparticles (LNPs) of two new pegylated lipopeptides for killing TNBCs (MDA-MB-231). The pegylated lipopeptides were synthesized by conjugating an n-hexadecyl hydrophobic tail to one end of a (PEG)27 unit the other distal end of which was covalently grafted with two previously reported tumor targeting RGDK- and CGKRK- peptides. The SEM images of the self-assembled LNPs formed upon dissolution of the pegylated lipopeptides in aqueous medium revealed formation of spherical aggregates. The degree of cellular uptake for the self-assembled LNPs formed by the pegylated CGKRK-lipopeptide were found to be significantly higher than that for the self-assembled LNPs formed by the pegylated RGDK-lipopeptide in MCF-7, MDA-MB-231, HEK-293 and HFF cells. Notably, about 60 % TNBCs (MDA-MB-231 cells) were killed upon treatment with commercially available potent JAK2 inhibitor (WP 1066) loaded LNPs of the pegylated RGDK-lipopeptide. Contrastingly, the same treatment killed only about 20 % non-cancerous HEK-293 cells. The self-assembled pegylated LNPs described herein open the door for undertaking preclinical studies in animal models for TNBCs.
Collapse
Affiliation(s)
- Wahida Rahaman
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Arabinda Chaudhuri
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| |
Collapse
|
24
|
Ilhami FB, Cahyaningrum SE, Wardana AP, Gultom NS, Subekti H, Rahmawati A, Puspitarini S. Meniran ( Phyllanthus niruri L.) embedded zeolitic imidazolate framework (ZIF-8) nanoparticle for cancer chemotherapy: supported molecular docking analysis. RSC Adv 2025; 15:223-230. [PMID: 39758894 PMCID: PMC11694506 DOI: 10.1039/d4ra06399f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025] Open
Abstract
Cancer is among the leading causes of mortality worldwide. Natural bioactive compounds like Meniran (Phyllanthus niruri L.) have been the focus of extensive research due to their potent anticancer properties. Nevertheless, drug delivery strategies may be necessary to encapsulate bioactive compounds, thereby reducing their toxicity and enhancing their stability. Herein, we successfully synthesized Meniran extract incorporated zeolitic imidazolate framework (ZIF-8) nanoparticles for anticancer therapy. Meniran-incorporated ZIF-8 nanoparticles possess unique advantages including well-distributed nanoparticles with rhombic dodecahedrons and excellent pH-responsiveness. In vitro analysis showed that Meniran-incorporated ZIF-8 nanoparticles have anticancer activity towards HeLa cells. Interestingly, computational simulations offer valuable insights into the molecular-level interaction mechanisms between ZIF-8 and specific proteins under cancer cells. As far as we are aware, this is the first report of natural bioactive compounds derived from Meniran encapsulated into nanoparticles as a drug delivery system, marking a significant advancement in the development of novel biomaterials for cancer treatment.
Collapse
Affiliation(s)
- Fasih Bintang Ilhami
- Department of Natural Science Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya Surabaya 60231 Indonesia
| | - Sari Edi Cahyaningrum
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Negeri Surabaya Surabaya 60231 Indonesia
| | - Andika Pramudya Wardana
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Negeri Surabaya Surabaya 60231 Indonesia
| | | | - Hasan Subekti
- Department of Natural Science Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya Surabaya 60231 Indonesia
| | - Astrid Rahmawati
- Department of Applied Chemistry, Osaka Institute of Technology Osaka 535-8585 Japan
| | - Sapti Puspitarini
- Department of Natural Science Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya Surabaya 60231 Indonesia
| |
Collapse
|
25
|
Meher N, Bidkar AP, Wadhwa A, Bobba KN, Dhrona S, Dasari C, Mu C, Fong COY, Cámara JA, Ali U, Basak M, Bulkley D, Steri V, Fontaine SD, Zhu J, Oskowitz A, Aggarwal RR, Sriram R, Chou J, Wilson DM, Seo Y, Santi DV, Ashley GW, VanBrocklin HF, Flavell RR. PET Imaging Using 89Zr-Labeled StarPEG Nanocarriers Reveals Heterogeneous Enhanced Permeability and Retention in Prostate Cancer. Mol Cancer Ther 2025; 24:141-151. [PMID: 39331510 DOI: 10.1158/1535-7163.mct-24-0024] [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: 01/15/2024] [Revised: 04/05/2024] [Accepted: 09/25/2024] [Indexed: 09/29/2024]
Abstract
The enhanced permeability and retention (EPR) effect controls passive nanodrug uptake in tumors and may provide a high tumor payload with prolonged retention for cancer treatment. However, EPR-mediated tumor uptake and distribution vary by cancer phenotype. Thus, we hypothesized that a companion PET imaging surrogate may benefit EPR-mediated therapeutic drug delivery. We developed two 89Zr-radiolabeled nanocarriers based on 4-armed starPEG40kDa with or without talazoparib (TLZ), a potent PARP inhibitor, as surrogates for the PEG-TLZ4 therapeutic scaffold. For PET imaging, PEG-DFB4 and PEG-DFB1-TLZ3 were radiolabeled with 89Zr by replacing one or all four copis of TLZ on PEG-TLZ4 with deferoxamine B (DFB). The radiolabeled nanodrugs [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 were tested in vivo in prostate cancer subcutaneous (s.c.) xenografts (22Rv1, LTL-545, and LTL-610) and 22Rv1 metastatic models. Their EPR-mediated tumoral uptake and penetration was compared with CT26, a known EPR-high cell line. MicroPET/CT images, organ biodistribution, and calculated kinetic parameters showed high uptake in CT26 and LTL-545 and moderate to low uptake in LTL-610 and 22Rv1. MicroPET/CT and high-resolution autoradiographic images showed nanocarrier penetration into highly permeable CT26, but heterogeneous peripheral accumulation was observed in LTL-545, LTL-610, and 22Rv1 s.c. xenografts and metastatic tumors. CD31 staining of tumor sections showed homogenous vascular development in CT26 tumors and heterogeneity in other xenografts. Both [89Zr]PEG-DFB4 and [89Zr]PEG-DFB1-TLZ3 showed similar accumulation and distribution in s.c. and metastatic tumor models. Both nanocarriers can measure tumor model passive uptake heterogeneity. Although heterogeneous, prostate cancer xenografts had low EPR. These starPEG nanocarriers could be used as PET imaging surrogates to predict drug delivery and efficacy.
Collapse
Affiliation(s)
- Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- National Institute of Pharmaceutical Education and Research, Lucknow, India
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Chandrashekhar Dasari
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Division of Vascular and Endovascular Surgery, University of California San Francisco, San Francisco, California
| | - Changhua Mu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Cyril O Y Fong
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Juan A Cámara
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Umama Ali
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Megha Basak
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - David Bulkley
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | - Jun Zhu
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - Adam Oskowitz
- Division of Vascular and Endovascular Surgery, University of California San Francisco, San Francisco, California
| | - Rahul R Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
| | - Jonathan Chou
- Department of Medicine, University of California San Francisco, San Francisco, California
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | | | | | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California
| |
Collapse
|
26
|
Serrano A, Casares N, Trocóniz IF, Lozano T, Lasarte JJ, Zalba S, Garrido MJ. Foxp3 inhibitory peptide encapsulated in a novel CD25-targeted nanoliposome promotes efficient tumor regression in mice. Acta Pharmacol Sin 2025; 46:171-183. [PMID: 39075226 PMCID: PMC11695603 DOI: 10.1038/s41401-024-01338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 06/06/2024] [Indexed: 07/31/2024]
Abstract
P60, a Foxp3 inhibitory peptide, can hinder the regulatory T cell (Treg) activity and impair tumor proliferation. However, low systemic stability and poor specificity have led to daily dosing to achieve therapeutic effect. Therefore, this study aims to improve P60 stability and specific delivery through its encapsulation in liposomes targeting CD25, constitutively expressed in Tregs. P60 liposomes formulated with DSPE-PEG750 or DSPE-PEG2000 were incubated with DSPE-PEG2000-Maleimide micelles conjugated to Fab' fragments of anti-CD25 to develop two targeted formulations or immunoliposomes (IL): IL-P602000 (DSPE-PEG2000 only) and IL-P60750 (combining DSPE-PEG750 and DSPE-PEG2000). P60 encapsulation efficiency was 50%-60% irrespective of PEG chain length. Treg uptake was 2.5 and 14 times higher for IL-PEG750 compared with IL-PEG2000 and non-targeted liposomes, respectively, in in-vitro assays. In fact, IL-P60750 allowed CD8+ T cells ex-vivo proliferation in presence of Treg at doses 10-20 times lower than for free P60. Antitumor response of P60 and IL-P60750 in monotherapy and combined with anti-PD-1 was evaluated in MC38 and LLCOVA tumor bearing mice. In MC38 model, IL-P60750 monotherapy induced total tumor regression in 40% of mice reaching 100% for anti-PD-1 combination. This effect was associated with a significant increase in activated CD8+ T cells in tumors. Notably, IL-P60750 also inhibited human Treg in ex-vivo assay, showing the translational capability of this formulation. In conclusion, IL-P60750 formulated with different PEG chain lengths, has demonstrated antitumor efficacy by selective inhibition of Treg activity and enhances the effect of anti-PD1. Altogether, this novel IL represents a promising nanoplatform for cancer immunotherapies.
Collapse
Affiliation(s)
- Alejandro Serrano
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Noelia Casares
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain
| | - Iñaki F Trocóniz
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
| | - Teresa Lozano
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain
| | - Juan J Lasarte
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain
- Program of Immunology and Immunotherapy, CIMA, Pamplona, Spain
| | - Sara Zalba
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain.
| | - María J Garrido
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdisNA), Pamplona, Spain.
| |
Collapse
|
27
|
Di Cintio F, Argenziano M, Scomparin A, Capolla S, Busato D, Steffè A, Mangogna A, Sblattero D, Cavalli R, Macor P, Dal Bo M, Toffoli G. The anti-glypican 1 AT101 antibody as targeting agent to effectively deliver chitosan nanobubbles to glioblastoma cells. Nanomedicine (Lond) 2025; 20:23-36. [PMID: 39620421 DOI: 10.1080/17435889.2024.2434451] [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: 08/09/2024] [Accepted: 11/22/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Recently, we developed AT101, an IgM-class mouse monoclonal antibody directed against glypican-1 (GPC1), a proteoglycan that can be considered as useful target for glioblastoma multiforme (GBM) treatment being specifically and highly expressed on GBM cell surface. Here, we proposed the use of AT101 as targeting agent in a drug delivery nanoplatfom to effectively deliver chitosan nanobubbles (NBs) for GBM treatment. METHODS Chitosan NBs were prepared and conjugated with AT101 or left unconjugated as control. RESULTS The ability of AT101 to bind the GPC1 protein was demonstrated by flow cytometry and immunofluorescence analysis in the "GBM-like" GPC1-expressing cell lines U-87 MG and T98G. AT101 was shown to bind GPC1-expressing GBM tumor samples by immunofluorescence. In-vivo experiments in the U-87 MG xenograft model showed that AT101 was able to bind GPC1 on cell surface and accumulate in U-87 MG tumor masses (p = 0.0002 respect to control). Moreover, in-vivo experiments showed that AT101 is able to target GPC1 when conjugated to chitosan NBs, thus increasing their specific deliver to GPC1-expressing cells of U-87 MG tumor, as compared to chitosan NBs not conjugated to AT101 (p = 0.02). CONCLUSIONS AT101 is an useful targeting agent for the development of drug delivery nanoplatforms for GBM treatment.
Collapse
Affiliation(s)
- Federica Di Cintio
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Anna Scomparin
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Sara Capolla
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Davide Busato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Aharon Steffè
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Alessandro Mangogna
- Department of Life Sciences, University of Trieste, Trieste, Italy
- Institute of Pathological Anatomy, Department of Medicine, University of Udine, Udine, Italy
| | | | - Roberta Cavalli
- Department of Drug Science and Technology, University of Turin, Turin, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
28
|
Dragowska WH, Singh J, Wehbe M, Anantha M, Edwards K, Gorski SM, Bally MB, Leung AWY. Liposomal Formulation of Hydroxychloroquine Can Inhibit Autophagy In Vivo. Pharmaceutics 2024; 17:42. [PMID: 39861690 PMCID: PMC11768354 DOI: 10.3390/pharmaceutics17010042] [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: 09/18/2024] [Revised: 11/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Preclinical studies have shown that the anti-malarial drug hydroxychloroquine (HCQ) improves the anti-cancer effects of various therapeutic agents by impairing autophagy. These findings are difficult to translate in vivo as reaching an effective HCQ concentration at the tumor site for extended times is challenging. Previously, we found that free HCQ in combination with gefitinib (Iressa®, ZD1839) significantly reduced tumor volume in immunocompromised mice bearing gefitinib-resistant JIMT-1 breast cancer xenografts. Here, we sought to evaluate whether a liposomal formulation of HCQ could effectively modulate autophagy in vivo and augment treatment outcomes in the same tumor model. Methods: We developed two liposomal formulations of HCQ: a pH-loaded formulation and a formulation based on copper complexation. The pharmacokinetics of each formulation was evaluated in CD1 mice following intravenous administration. An efficacy study was performed in immunocompromised mice bearing established JIMT-1tumors. Autophagy markers in tumor tissue harvested after four weeks of treatment were assessed by Western blot. Results: The liposomal formulations engendered ~850-fold increases in total drug exposure over time relative to the free drug. Both liposomal and free HCQ in combination with gefitinib provided comparable therapeutic benefits (p > 0.05). An analysis of JIMT-1 tumor tissue indicated that the liposomal HCQ and gefitinib combination augmented the inhibition of autophagy in vivo compared to the free HCQ and gefitinib combination as demonstrated by increased LC3-II and p62/SQSTM1 (p62) protein levels. Conclusions: The results suggest that liposomal HCQ has a greater potential to modulate autophagy in vivo compared to free HCQ; however, this did not translate to better therapeutic effects when used in combination with gefitinib to treat a gefitinib-resistant tumor model.
Collapse
Affiliation(s)
- Wieslawa H. Dragowska
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (W.H.D.); (M.A.); (M.B.B.)
| | - Jagbir Singh
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (W.H.D.); (M.A.); (M.B.B.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Mohamed Wehbe
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (W.H.D.); (M.A.); (M.B.B.)
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Malathi Anantha
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (W.H.D.); (M.A.); (M.B.B.)
| | - Katarina Edwards
- Department of Chemistry, Ångström Laboratory, Uppsala University, 751 20 Uppsala, Sweden;
| | - Sharon M. Gorski
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada;
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Vancouver, BC V5A 1S6, Canada
| | - Marcel B. Bally
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (W.H.D.); (M.A.); (M.B.B.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6T 1Z3, Canada
| | - Ada W. Y. Leung
- Department of Experimental Therapeutics, BC Cancer, Vancouver, BC V5Z 1L3, Canada; (W.H.D.); (M.A.); (M.B.B.)
- Cuprous Pharmaceuticals Inc., Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
29
|
Wang K, Geng S, Wang F, Fang B, Qian H, Li Y, Zhou Y, Chen Y, Yu Z. Natural epigallocatechin-3-gallocarboxylate nanoformulation loaded doxorubicin to construct a novel and low cardiotoxicity chemotherapeutic drug for high-efficiency breast cancer therapy. J Nanobiotechnology 2024; 22:793. [PMID: 39719646 DOI: 10.1186/s12951-024-03069-0] [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: 09/09/2024] [Accepted: 12/10/2024] [Indexed: 12/26/2024] Open
Abstract
Anthracycline doxorubicin (DOX) remains the first-line chemotherapeutic drug for the efficient treatment of breast cancer, but its severe cardiotoxicity limits its long-term application in clinical tumor chemotherapy. Until now, the pathogenesis mechanism of DOX-induced cardiotoxicity (DIC) is still not fully understood. According to current studies, the oxidative stress caused by the imbalance of reactive oxygen species (ROS) and reactive nitrogen species (RNS) production and mitochondrial dysfunction in myocardial cells are closely related to DIC. Presently, the usual technology to solve the DIC problem is to use a multifunctional nanoplatform to load DOX and obtain a new medicinal agent, thereby enhancing the efficacy of chemotherapeutic drugs and reducing toxic side effects.Herein, the present investigation employed the Mannich condensation reaction, initiated by L-cysteine and (-)-epigallocatechin-3-gallocarboxylate (EGCG), to synthesize EGCG&Cys nanoformulation with both anti-tumor and anti-oxidant properties. The EGCG&Cys were then employed as the DOX carrier to construct a novel chemotherapeutic drug, EGCG&Cys(DOX), for high-efficiency breast cancer treatment. The tumor growth inhibition index of EGCG&Cys(DOX) in tumor-bearing mice was 12.56% superior to the DOX group with the same concentration. Meanwhile, the anti-oxidant properties of EGCG can effectively eliminate a large amount of free radicals produced by DOX and significantly alleviate DIC by improving mitochondrial functional pathways. Ultrasound echocardiography (UCG) showed that the mean LVEF and LEFS values in the 5 mg/kg DOX treatment group were significantly reduced by 54.4% and 63.4%, and the EGCG&Cys(DOX) group mice were consistent with those of the non-chemotherapy group. Moreover, electrocardiogram, serum biochemical indicators, and histopathological analysis results also demonstrate that the cardiotoxicity of EGCG&Cys(DOX) novel chemotherapy drugs is significantly reduced. Consequently, this study presents a new technology for preparing highly efficient and safe nano-chemotherapeutic drugs and an in-depth evaluation of the antitumor efficacy and safety of the synthesized novel drugs, which gave fresh life to the development of nanomedicine in the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Ke Wang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Siqi Geng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Fang Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Baoru Fang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Huifeng Qian
- Department of Clinical Laboratory, Shaoxing Second Hospital, Shaoxing, 312000, Zhejiang, China
| | - Ying Li
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Yiqing Zhou
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Yanping Chen
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Zhangsen Yu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China.
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
30
|
Velapure P, Kansal D, Bobade C. Tumor microenvironment-responsive nanoformulations for breast cancer. DISCOVER NANO 2024; 19:212. [PMID: 39708097 DOI: 10.1186/s11671-024-04122-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/07/2024] [Indexed: 12/23/2024]
Abstract
Nanomedicine, the most promising approach for regulated and targeted drug delivery, is frequently applied in cancer treatment. Essentially, accumulating evidence indicates that nanomedicine has positive results in the treatment of breast cancer (BC), with many BC patients benefiting from nanomedicine-related treatments. Currently, nanodrug delivery systems based on stimulus responses are gaining popularity because of their additional ability to manage drug release depending on the interior environment of the cancer. This review includes a synopsis of several types of internal (pH, redox, enzyme, reactive oxygen species, and hypoxia) stimuli-responsive nanoparticle drug delivery systems as well as perspectives for forthcoming times. Stimulus-responsive nanoparticles can remain stable under physiological conditions while being rapidly activated to release drugs in response to specific stimuli, prolonging blood circulation and increasing cancer cellular uptake, resulting in excellent therapeutic performance and improved biosafety. In this paper, we discuss tumor microenvironment responsive Nanoformulation for breast cancer treatment.
Collapse
Affiliation(s)
- Pallavi Velapure
- School of Health Science and Technology, Dr. Vishwanath Karad MIT World Peace University, S.No. 124, MIT Campus, Paud Road, Kothrud, Pune, 411038, Maharashtra, India
| | - Divyanshi Kansal
- School of Health Science and Technology, Dr. Vishwanath Karad MIT World Peace University, S.No. 124, MIT Campus, Paud Road, Kothrud, Pune, 411038, Maharashtra, India
| | - Chandrashekhar Bobade
- School of Health Science and Technology, Dr. Vishwanath Karad MIT World Peace University, S.No. 124, MIT Campus, Paud Road, Kothrud, Pune, 411038, Maharashtra, India.
| |
Collapse
|
31
|
Roussel T, Cruz-Dubois T, Louis B, Laurini E, Ding L, Balasse L, Nail V, Dignat-George F, Giorgio S, Pricl S, Guillet B, Garrigue P, Peng L. Impact of inner hydrophobicity of dendrimer nanomicelles on biodistribution: a PET imaging study. J Mater Chem B 2024. [PMID: 39699216 DOI: 10.1039/d4tb01266f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Self-assembly is a powerful strategy for building nanosystems for biomedical applications. We have recently developed small amphiphilic dendrimers capable of self-assembling into nanomicelles for tumor imaging. In this context, we studied the impact of increased hydrophobicity of the amphiphilic dendrimer on hydrophilic/hydrophobic balance and consequently on the self-assembly and subsequent biodistribution. Remarkably, despite maintaining the exact same surface chemistry, similar zeta potential, and small size, the altered and enlarged hydrophobic component within the amphiphilic dendrimer led to enhanced stability of the self-assembled nanomicelles, with prolonged circulation time and massive accumulation in the liver. This study reveals that even structural alteration within the interior of nanomicelles can dramatically impact biodistribution profiles. This finding highlights the deeper complexity of rational design for nanomedicine and the need to consider factors other than surface charge and chemistry, as well as size, all of which significantly impact the biodistribution of self-assembling nanosystems.
Collapse
Affiliation(s)
- Tom Roussel
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| | - Twiany Cruz-Dubois
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Beatrice Louis
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Erik Laurini
- Molecular Biology and Nanotechnology Laboratory, Department of Engineering and Architectures, University of Trieste, Trieste 34127, Italy
| | - Ling Ding
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| | - Laure Balasse
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Vincent Nail
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | | | - Suzanne Giorgio
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| | - Sabrina Pricl
- Molecular Biology and Nanotechnology Laboratory, Department of Engineering and Architectures, University of Trieste, Trieste 34127, Italy
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz 90-136, Poland
| | - Benjamin Guillet
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Philippe Garrigue
- Aix Marseille University, INSERM, INRAE, C2VN, Marseille, France
- Aix Marseille University, CNRS, CERIMED, Marseille, France
| | - Ling Peng
- Aix Marseille University, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille (CINaM), UMR 7325, Equipe Labellisée Ligue Contre le Cancer, Marseille, France.
| |
Collapse
|
32
|
Yin M, Zhang X, Zhang T, Bao Z, He Z. Folic Acid-Targeted Mixed Pluronic Micelles for Delivery of Triptolide. Polymers (Basel) 2024; 16:3485. [PMID: 39771337 PMCID: PMC11677570 DOI: 10.3390/polym16243485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The present study aimed to explore an ideal delivery system for triptolide (TPL) by utilizing the thin-film hydration method to prepare drug-loaded, folate-modified mixed pluronic micelles (FA-F-127/F-68-TPL). Scanning electron microscopy and atomic force microscopy showed that the drug-loaded micelles had a spherical shape with a small particle size, with an average of 30.7 nm. Cell viability experiments showed that FA-F-127/F-68-TPL significantly reduced HepG2 cell viability, exhibiting strong cytotoxicity. Its cytotoxicity was markedly enhanced compared with bare TPL. Nile red (Nr) was used as a model drug to prepare FA-F-127/F-68-Nr to further validate its tumor-targeting and cellular uptake capability. After coincubation with HepG2 cells, a multifunctional microplate reader showed that intracellular fluorescence intensity significantly increased, indicating that FA-F-127/F-68-Nr could more effectively enter the cells. A nude mouse model of subcutaneous hepatocellular carcinoma was constructed. Following tail vein injection of FA-F-127/F-68-Nr, the fluorescence imaging system showed that FA-F127/F-68-Nr could significantly target tumor tissue, and even if entering the small-sized tumor was challenging, it could be excreted through urine. Nude mice with subcutaneous hepatocellular carcinoma were treated with tail vein injections of FA-F-127/F-68-TPL (45 µg/kg) every other day for 21 days. The results showed that the growth of the transplanted tumors was significantly slowed, with no significant difference compared with bare TPL. In summary, the FA-F-127/F-68-TPL exhibits the advantages of low cost, excellent biological properties, active/passive targeting capabilities, notable cytotoxicity against liver cancer cells, and significant inhibition of transplanted hepatocellular carcinoma growth. Significantly, the FA-F-127/F-68-TPL, despite challenges in targeting tumors with an insignificant EPR effect, can be efficiently excreted via the kidneys, thereby preventing the release of the drug during prolonged circulation and potential damage to normal tissues. Therefore, FA-F-127/F-68-TPL represents a promising antitumor drug delivery system.
Collapse
Affiliation(s)
- Meizhen Yin
- Medical College, Inner Mongolia Minzu University, Tongliao 028043, China
| | | | | | | | | |
Collapse
|
33
|
Wang Y, Ma K, Kang M, Yan D, Niu N, Yan S, Sun P, Zhang L, Sun L, Wang D, Tan H, Tang BZ. A new era of cancer phototherapy: mechanisms and applications. Chem Soc Rev 2024; 53:12014-12042. [PMID: 39494674 DOI: 10.1039/d4cs00708e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
The past decades have witnessed great strides in phototherapy as an experimental option or regulation-approved treatment in numerous cancer indications. Of particular interest is nanoscale photosensitizer-based phototherapy, which has been established as a prominent candidate for advanced tumor treatment by virtue of its high efficacy and safety. Despite considerable research progress on materials, methods and devices in nanoscale photosensitizing agent-based phototherapy, their mechanisms of action are not always clear, which impedes their practical application in cancer treatment. Hence, from a new perspective, this review elaborates the working mechanisms, involving impairment and moderation effects, of diverse phototherapies on cells, organelles, organs, and tissues. Furthermore, the most current available phototherapy modalities are categorized as photodynamic, photothermal, photo-immune, photo-gas, and radio therapies in this review. A comprehensive understanding of the inferiority and superiority of various phototherapies will facilitate the advent of a new era of cancer phototherapy.
Collapse
Affiliation(s)
- Yuanwei Wang
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Ke Ma
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Miaomiao Kang
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Dingyuan Yan
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Niu Niu
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Saisai Yan
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Panpan Sun
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Luzhi Zhang
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Lijie Sun
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Dong Wang
- Center for AIE Research, Guangdong Provincial Key Laboratory of New Energy Materials Service Safety, College of Materials Science and Engineering Shenzhen University, Shenzhen 518060, P. R. China.
| | - Hui Tan
- Center for Child Care and Mental Health (CCCMH) Shenzhen Children's Hospital, Shenzhen 518026, P. R. China.
| | - Ben Zhong Tang
- School of Science and Engineering, Shenzhen Institute of Aggregate Science and Technology, The Chinese University of Hong Kong Shenzhen, (CUHK-Shenzhen), Guangdong 518172, China.
| |
Collapse
|
34
|
Cizkova J, Dolezal OJ, Buchta V, Pospichal J, Blanar V, Sinkorova Z, Carrillo A. Golden era of radiosensitizers. Front Vet Sci 2024; 11:1450776. [PMID: 39711799 PMCID: PMC11659289 DOI: 10.3389/fvets.2024.1450776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/21/2024] [Indexed: 12/24/2024] Open
Abstract
The past 30 years have brought undeniable progress in medicine, biology, physics, and research. Knowledge of the nature of the human body, diseases, and disorders has been constantly improving, and the same is true regarding their treatment and diagnosis. One of the greatest advances in recent years has been the introduction of nanoparticles (NPs) into medicine. NPs refer to a material at a nanometer scale (0.1-100 nm) with features (specific physical, chemical, and biological properties) that are broadly and increasingly used in the medical field. Their applications in cancer treatment and radiotherapy seem particularly attractive. In this field, inorganic/metal NPs with high atomic number Z have been employed mainly due to their ability to enhance ionizing radiation's photoelectric and Compton effects and thereby increase conventional radiation therapy's efficacy. The improvement NPs enable relates to their enhanced permeation ability and longer retention effect in tumor cells, capacity to reduce toxicity of commercially available cancer drugs through advanced NPs drug delivery systems, radiation sensitizers of tumors, or enhancers of radiation doses to tumors. Advanced options according to size, core, and surface modification allow even such multimodal approaches in therapy as nanotheranostics or combined treatments. The current state of knowledge emphasizes the role of gold nanoparticles (AuNPs) in sensitizing tumors to radiation. We have reviewed AuNPs and their radiosensitizing power during radiation treatment. Our results are divided into groups based on AuNPs' surface modification and/or core structure design. This study provides a complete summary of the in vivo sensitizing effect of AuNPs, surface-modified AuNPs, and AuNPs combined with different elements, providing evidence for further successful veterinarian and clinical implementation.
Collapse
Affiliation(s)
- Jana Cizkova
- Department of Radiobiology, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czechia
| | - Ondrej Jan Dolezal
- Department of Radiobiology, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czechia
| | - Vojtech Buchta
- Department of Clinical Subspecialties, Faculty of Health Studies, University of Pardubice, Pardubice, Czechia
| | - Jan Pospichal
- Department of Clinical Subspecialties, Faculty of Health Studies, University of Pardubice, Pardubice, Czechia
| | - Vit Blanar
- Department of Nursing, Faculty of Health Studies, University of Pardubice, Pardubice, Czechia
| | - Zuzana Sinkorova
- Department of Radiobiology, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czechia
| | - Anna Carrillo
- Department of Radiobiology, Military Faculty of Medicine, University of Defence, Hradec Kralove, Czechia
| |
Collapse
|
35
|
Cui Y, Yang K, Guo C, Xia Z, Jiang B, Xue Y, Song B, Hu W, Zhang M, Wei Y, Zhang C, Zhang S, Fang J. Carbon monoxide as a negative feedback mechanism on HIF-1α in the progression of metabolic-associated fatty liver disease. Nitric Oxide 2024; 153:1-12. [PMID: 39369813 DOI: 10.1016/j.niox.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) encompasses various chronic liver conditions, yet lacks approved drugs. Hypoxia-inducible factor-1α (HIF-1α) is pivotal in MAFLD development. Our prior research highlighted the efficacy of the nano-designed carbon monoxide (CO) donor, targeting HIF-1α in a mouse hepatic steatosis model. Given heme oxygenase-1 (HO-1, a major downstream molecule of HIF-1α) as the primary source of intrinsic CO, we hypothesized that upregulation of HO-1/CO, responsive to HIF-1α, forms a negative feedback loop regulating MAFLD progression. In this study, we explored the potential negative feedback mechanism of CO on HIF-1α and its downstream effects on MAFLD advancement. HIF-1α emerges early in hepatic steatosis induced by a high-fat (HF) diet, triggering increased HO-1 and inflammation. SMA/CORM2 effectively suppresses HIF-1α and steatosis progression when administered within the initial week of HF diet initiation but loses impact later. In adipose tissues, concurrent metabolic dysfunction and inflammation with HIF-1α activation suggest adipose tissue expansion initiates HF-induced steatosis, triggering hypoxia and liver inflammation. Notably, in an in vitro study using mouse hepatocytes treated with fatty acids, downregulating HO-1 intensified HIF-1α induction at moderate fatty acid concentrations. However, this effect diminished at high concentrations. These results suggest the HIF-1α-HO-1-CO axis as a feedback loop under physiological and mild pathological conditions. Excessive HIF-1α upregulation in pathological conditions overwhelms the CO feedback loop. Additional CO application effectively suppresses HIF-1α and disease progression, indicating potential application for MAFLD control.
Collapse
Affiliation(s)
- Yingying Cui
- Peking University First Hospital Ningxia Women and Children's Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, 750000, China; Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Kai Yang
- Department of Medical Technology, Anhui Medical College, No.632, Furong Road, Hefei, Anhui Province, China
| | - Chunyu Guo
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Zhengmei Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Benchun Jiang
- Department of Gastricintestinal Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanni Xue
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Bingdong Song
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Weirong Hu
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Mingjie Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, 110004, Liaoning, China
| | - Yanyan Wei
- Department of Infectious Disease, the First Affiliated Hospital of Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Cheng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China
| | - Shichen Zhang
- Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China.
| | - Jun Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, No 81 Meishan Road, Hefei, 230022, Anhui, China; Anhui Provincial Center for Maternal and Child Health Genetics, School of Public Health and Health Management, Anhui Medical College, No 632 Furong Road, Hefei, 230601, Anhui, China; Faculty of Pharmaceutical Sciences, Sojo University, Ikeda 4-22-1, Kumamoto, 860-0082, Japan.
| |
Collapse
|
36
|
Carigga Gutierrez NM, Clainche TL, Bulin A, Leo S, Kadri M, Abdelhamid AGA, Pujol‐Solé N, Obaid G, Hograindleur M, Gardette V, Busser B, Motto‐Ros V, Josserand V, Henry M, Sancey L, Hurbin A, Elleaume H, Kandiah E, Guével XL, Coll J, Broekgaarden M. Engineering Radiocatalytic Nanoliposomes with Hydrophobic Gold Nanoclusters for Radiotherapy Enhancement. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404605. [PMID: 39473330 PMCID: PMC11636064 DOI: 10.1002/adma.202404605] [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: 03/29/2024] [Revised: 09/21/2024] [Indexed: 12/13/2024]
Abstract
Chemoradiation therapy is on the forefront of pancreatic cancer care, and there is a continued effort to improve its safety and efficacy. Liposomes are widely used to improve chemotherapy safety, and may accurately deliver high-Z element- radiocatalytic nanomaterials to cancer tissues. In this study, the interaction between X-rays and long-circulating nanoliposome formulations loaded with gold nanoclusters is explored in the context of oxaliplatin chemotherapy for desmoplastic pancreatic cancer. Hydrophobic gold nanoclusters stabilized with dodecanethiol (AuDDT) are efficiently incorporated in nanoliposomal bilayers. AuDDT-nanoliposomes significantly augmented radiation-induced •OH production, which is most effective with monochromatic X-rays at energies that exceed the K-shell electron binding energy of Au (81.7 keV). Cargo release assays reveal that AuDDT-nanoliposomes can permeabilize lipid bilayers in an X-ray dose- and formulation-dependent manner. The radiocatalytic effect of AuDDT-nanoliposomes significantly augments radiotherapy and oxaliplatin-chemoradiotherapy outcomes in 3D pancreatic microtumors. The PEGylated AuDDT-nanoliposomes display high tumor accumulation in an orthotopic mouse model of pancreatic cancer, showing promise for nanoliposomes as carriers for radiocatalytic nanomaterials. Altogether, compelling proof for chemo-radiation dose-enhancement using AuDDT-nanoliposomes is presented. Further improving the nanoliposomal loading of high-Z elements will advance the safety, efficacy, and translatability of such chemoradiation dose-enhancement approaches.
Collapse
Affiliation(s)
| | - Tristan Le Clainche
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Anne‐Laure Bulin
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Sofia Leo
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
- Porphychem SASLongvic21600France
| | - Malika Kadri
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Ahmed Gamal Ali Abdelhamid
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Núria Pujol‐Solé
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Girgis Obaid
- Department of BioengineeringUniversity of Texas at DallasRichardsonTX75080USA
| | - Marc‐André Hograindleur
- European Synchrotron Radiation FacilityCM01 Beamline71 Avenue des MartyrsGrenoble38000France
| | - Vincent Gardette
- Université Lyon 1Institut Lumière Matière, CNRS UMR 5306Université de LyonVilleurbanneFrance
- University Hospital of Grenoble AlpesGrenoble38000France
| | - Benoit Busser
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
- University Hospital of Grenoble AlpesGrenoble38000France
| | - Vincent Motto‐Ros
- Université Lyon 1Institut Lumière Matière, CNRS UMR 5306Université de LyonVilleurbanneFrance
- University Hospital of Grenoble AlpesGrenoble38000France
| | - Véronique Josserand
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Maxime Henry
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Lucie Sancey
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Amandine Hurbin
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Hélène Elleaume
- Université de Grenoble‐AlpesSynchrotron Radiation for Biomedicine, Inserm UA072280 Rue de la PiscineSaint Martin d'Hères38400France
| | - Eaazhisai Kandiah
- Department of BioengineeringUniversity of Texas at DallasRichardsonTX75080USA
| | - Xavier Le Guével
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Jean‐Luc Coll
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| | - Mans Broekgaarden
- Université Grenoble‐AlpesInserm U1209CNRS UMR 5309Institute for Advanced BiosciencesAllée des AlpesLa Tronche38700France
| |
Collapse
|
37
|
Keshavarz Shahbaz S, Koushki K, Izadi O, Penson PE, Sukhorukov VN, Kesharwani P, Sahebkar A. Advancements in curcumin-loaded PLGA nanoparticle delivery systems: progressive strategies in cancer therapy. J Drug Target 2024; 32:1207-1232. [PMID: 39106154 DOI: 10.1080/1061186x.2024.2389892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Cancer is a leading cause of death worldwide, and imposes a substantial socioeconomic burden with little impact especially on aggressive types of cancer. Conventional therapies have many serious side effects including generalised systemic toxicity which limits their long-term use. Tumour resistance and recurrence is another main problem associated with conventional therapy. Purified or extracted natural products have been investigated as cost-effective cancer chemoprotective agents with the potential to reverse or delaying carcinogenesis. Curcumin (CUR) as a natural polyphenolic component, exhibits many pharmacological activities such as anti-cancer, anti-inflammatory, anti-microbial, activity against neurodegenerative diseases including Alzheimer, antidiabetic activities (type II diabetes), anticoagulant properties, wound healing effects in both preclinical and clinical studies. Despite these effective protective properties, CUR has several limitations, including poor aqueous solubility, low bioavailability, chemical instability, rapid metabolism and a short half-life time. To overcome the pharmaceutical problems associated with free CUR, novel nanomedicine strategies (including polymeric nanoparticles (NPs) such as poly (lactic-co-glycolic acid) (PLGA) NPs have been developed. These formulations have the potential to improve the therapeutic efficacy of curcuminoids. In this review, we comprehensively summarise and discuss recent in vitro and in vivo studies to explore the pharmaceutical significance and clinical benefits of PLGA-NPs delivery system to improve the efficacy of CUR in the treatment of cancer.
Collapse
Affiliation(s)
- Sanaz Keshavarz Shahbaz
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-Communicable Disease, Qazvin University of Medical Sciences, Qazvin, Iran
- USERN Office, Qazvin University of Medical Science, Qazvin, Iran
| | - Khadijeh Koushki
- Department of Neurosurgery, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Omid Izadi
- Department of Industrial Engineering, ACECR Institute of Higher Education Kermanshah, Kermanshah, Iran
| | - Peter E Penson
- Clinical Pharmacy and Therapeutics Research Group, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
- Liverpool Centre for Cardiovascular Science, Liverpool, UK
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Centre, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
38
|
Han MM, Fan YK, Zhang Y, Dong ZQ. Advances in herbal polysaccharides-based nano-drug delivery systems for cancer immunotherapy. J Drug Target 2024; 32:311-324. [PMID: 38269853 DOI: 10.1080/1061186x.2024.2309661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/20/2024] [Indexed: 01/26/2024]
Abstract
The boom in cancer immunotherapy has provided many patients with a better chance of survival, but opportunities often come with challenges. Single immunotherapy is not good enough to eradicate tumours, and often fails to achieve the desired therapeutic effect because of the low targeting of immunotherapy drugs, and causes more side effects. As a solution to this problem, researchers have developed several nano Drug Delivery Systems (NDDS) to deliver immunotherapeutic agents to achieve good therapeutic outcomes. However, traditional drug delivery systems (DDS) have disadvantages such as poor bioavailability, high cytotoxicity, and difficulty in synthesis, etc. Herbal Polysaccharides (HPS), derived from natural Chinese herbs, inherently possess low toxicity. Furthermore, the biocompatibility, biodegradability, hydrophilicity, ease of modification, and immunomodulatory activities of HPS offer unique advantages in substituting traditional DDS. This review initially addresses the current developments and challenges in immunotherapy. Subsequently, it focuses on the immunomodulatory mechanisms of HPS and their design as nanomedicines for targeted drug delivery in tumour immunotherapy. Our findings reveal that HPS-based nanomedicines exhibit significant potential in enhancing the efficacy of cancer immunotherapy, providing crucial theoretical foundations and practical guidelines for future clinical applications.
Collapse
Affiliation(s)
- Miao-Miao Han
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yi-Kai Fan
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Zhang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| | - Zheng-Qi Dong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine from Ministry of Education, Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription from Chinese Academy of Medical Sciences, Department of Pharmaceutics, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
- Joint Research Center for Chinese Medicinal Herbs, IMPLAD, ABRC & ACCL, Beijing, China
| |
Collapse
|
39
|
Liu L, Yang M, Chen Z. Surface functionalized nanomaterial systems for targeted therapy of endocrine related tumors: a review of recent advancements. Drug Deliv 2024; 31:2390022. [PMID: 39138394 PMCID: PMC11328606 DOI: 10.1080/10717544.2024.2390022] [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: 03/19/2024] [Revised: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 08/15/2024] Open
Abstract
The application of multidisciplinary techniques in the management of endocrine-related cancers is crucial for harnessing the advantages of multiple disciplines and their coordinated efforts in eliminating tumors. Due to the malignant characteristics of cancer cells, they possess the capacity to develop resistance to traditional treatments such as chemotherapy and radiotherapy. Nevertheless, despite diligent endeavors to enhance the prediction of outcomes, the overall survival rate for individuals afflicted with endocrine-related malignancy remains quite miserable. Hence, it is imperative to investigate innovative therapy strategies. The latest advancements in therapeutic tactics have offered novel approaches for the therapy of various endocrine tumors. This paper examines the advancements in nano-drug delivery techniques and the utilization of nanomaterials for precise cancer cures through targeted therapy. This review provides a thorough analysis of the potential of combined drug delivery strategies in the treatment of thyroid cancer, adrenal gland tumors, and pancreatic cancer. The objective of this study is to gain a deeper understanding of current therapeutic approaches, stimulate the development of new drug DDS, and improve the effectiveness of treatment for patients with these diseases. The intracellular uptake of pharmaceuticals into cancer cells can be significantly improved through the implantation of synthetic or natural substances into nanoparticles, resulting in a substantial reduction in the development of endocrine malignancies.
Collapse
Affiliation(s)
- Limei Liu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Miao Yang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ziyang Chen
- Department of Gastroenterology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
40
|
Chen J, Yao Y, Mao X, Chen Y, Ni F. Liver-targeted delivery based on prodrug: passive and active approaches. J Drug Target 2024; 32:1155-1168. [PMID: 39072411 DOI: 10.1080/1061186x.2024.2386416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND The liver, a central organ in human metabolism, is often the primary target for drugs. However, conditions such as viral hepatitis, cirrhosis, non-alcoholic fatty liver disease (NAFLD), and hepatocellular carcinoma (HCC) present substantial health challenges worldwide. Existing treatments, which suffer from the non-specific distribution of drugs, frequently fail to achieve desired efficacy and safety, risking unnecessary liver harm and systemic side effects. PURPOSE The aim of this review is to synthesise the latest progress in the design of liver-targeted prodrugs, with a focus on passive and active targeting strategies, providing new insights into the development of liver-targeted therapeutic approaches. METHODS This study conducted an extensive literature search through databases like Google Scholar, PubMed, Web of Science, and China National Knowledge Infrastructure (CNKI), systematically collecting and selecting recent research on liver-targeted prodrugs. The focus was on targeting mechanisms, including the Enhanced Permeability and Retention (EPR) effect, the unique microenvironment of liver cancer, and active targeting through specific transporters and receptors. RESULTS Active targeting strategies achieve precise drug delivery by binding specific ligands to liver surface receptors. Passive targeting takes advantage of the EPR effect and tumour characteristics to enrich drugs in liver tumours. The review details successful cases of using small molecule ligands, peptides, antibodies and nanoparticles as drug carriers. CONCLUSION Liver-targeted prodrug strategies show great potential in enhancing the efficacy of drug treatment and reducing side effects for liver diseases. Future research should balance the advantages and limitations of both targeting strategies, focusing on optimising drug design and targeting efficiency, especially for clinical application. In-depth research on liver-specific receptors and the development of innovative targeting molecules are crucial for advancing the field of liver-targeted prodrugs.
Collapse
Affiliation(s)
- Jiaqi Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingrui Yao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoran Mao
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuzhou Chen
- Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Feng Ni
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| |
Collapse
|
41
|
Schäfer M, Hildenbrand G, Hausmann M. Impact of Gold Nanoparticles and Ionizing Radiation on Whole Chromatin Organization as Detected by Single-Molecule Localization Microscopy. Int J Mol Sci 2024; 25:12843. [PMID: 39684554 DOI: 10.3390/ijms252312843] [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: 10/15/2024] [Revised: 11/24/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
In radiation tumor therapy, irradiation, on one hand, should cause cell death to the tumor. On the other hand, the surrounding non-tumor tissue should be maintained unaffected. Therefore, methods of local dose enhancements are highly interesting. Gold nanoparticles, which are preferentially uptaken by very-fast-proliferating tumor cells, may enhance damaging. However, the results in the literature obtained from cell culture and animal tissue experiments are very contradictory, i.e., only some experiments reveal increased cell killing but others do not. Thus, a better understanding of cellular mechanisms is required. Using the breast cancer cell model SkBr3, the effects of gold nanoparticles in combination with ionizing radiation on chromatin network organization were investigated by Single-Molecule Localization Microscopy (SMLM) and applications of mathematical topology calculations (e.g., Persistent Homology, Principal Component Analysis, etc.). The data reveal a dose and nanoparticle dependent re-organization of chromatin, although colony forming assays do not show a significant reduction of cell survival after the application of gold nanoparticles to the cells. In addition, the spatial organization of γH2AX clusters was elucidated, and characteristic changes were obtained depending on dose and gold nanoparticle application. The results indicate a complex response of ALU-related chromatin and heterochromatin organization correlating to ionizing radiation and gold nanoparticle incorporation. Such complex whole chromatin re-organization is usually associated with changes in genome function and supports the hypothesis that, with the application of gold nanoparticles, not only is DNA damage increasing but also the efficiency of DNA repair may be increased. The understanding of complex chromatin responses might help to improve the gold nanoparticle efficiency in radiation treatment.
Collapse
Affiliation(s)
- Myriam Schäfer
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
- Faculty of Engineering, University of Applied Sciences Aschaffenburg, Würzburger Str. 45, 63743 Aschaffenburg, Germany
| | - Georg Hildenbrand
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
- Faculty of Engineering, University of Applied Sciences Aschaffenburg, Würzburger Str. 45, 63743 Aschaffenburg, Germany
| | - Michael Hausmann
- Kirchhoff-Institute for Physics, Heidelberg University, Im Neuenheimer Feld 227, 69120 Heidelberg, Germany
| |
Collapse
|
42
|
Wang D, Qiu CJ, Chu Y, Zhang A, Huang R, Pan SJ, Tan L. A Polymeric Vesicle System for Combined Lung Cancer Therapy through Chemotherapy and Vasculature Normalization. Biomater Res 2024; 28:0117. [PMID: 39606153 PMCID: PMC11599482 DOI: 10.34133/bmr.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Lung cancer remains a great threat to human health despite the rapid development of various therapeutic methods. Chemotherapy continues to be the most commonly employed treatment for lung cancer; however, it often suffers from low drug delivery efficiency and severe side effects. To enhance the therapeutic efficacy of chemotherapy, we developed a novel strategy that integrates tumor vasculature normalization with the co-delivery of therapeutic agents. This strategy employs a diblock polymeric vesicle with a reduction-sensitive linkage. Paclitaxel (PTX) is encapsulated in the bilayer, while an acid-sensitive nitric oxide (NO) precursor, DETA NONOate, and zinc oxide nanoparticles (ZnO NPs) are loaded into the central cavity. The resulting nanosystem, (ZnO,NONO)@Ves-PTX, is designed to release NO under the acidic conditions typical of the tumor microenvironment (TME) and intracellular environment. The released NO in the TME inhibits angiogenesis, thereby facilitating the delivery and distribution of therapeutic agents. Upon internalization by tumor cells, (ZnO,NONO)@Ves-PTX decomposes in response to intracellular glutathione (GSH), releasing the loaded agents. DETA NONOate and ZnO NPs generate NO and Zn2+ ions, respectively, at the intracellular pH, which synergistically inhibit tumor growth alongside PTX. This combined therapeutic approach demonstrated remarkable potential in improving the chemotherapeutic efficacy for lung cancer, offering a promising direction for future cancer treatments.
Collapse
Affiliation(s)
- Ding Wang
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
| | - Cheng-Jie Qiu
- Department of Neurosurgery, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yaoqing Chu
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
| | - Anzhuo Zhang
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
| | - Ran Huang
- Academy for Engineering and Technology; Yiwu Research Institute; Zhuhai Fudan Innovation Institute,
Fudan University, Shanghai 200433, China
- Center for Innovation and Entrepreneurship,
Taizhou Institute of Zhejiang University, Taizhou, Zhejiang 318000, China
| | - Si-Jian Pan
- Department of Neurosurgery, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lianjiang Tan
- School of Materials Science and Engineering,
Shanghai Institute of Technology, Shanghai 201418, China
- Department of Neurosurgery, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
43
|
Sorroza-Martínez K, González-Sánchez I, Villamil-Ramos R, Cerbón M, Guerrero-Álvarez JA, Coronel-Cruz C, Rivera E, González-Méndez I. Using Poly(amidoamine) PAMAM-βCD Dendrimer for Controlled and Prolonged Delivery of Doxorubicin as Alternative System for Cancer Treatment. Pharmaceutics 2024; 16:1509. [PMID: 39771488 PMCID: PMC11728618 DOI: 10.3390/pharmaceutics16121509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: Doxorubicin (Dox) is an anticancer drug used in the treatment of a wide range of solid tumors; however, Dox causes systemic toxicity and irreversible cardiotoxicity. The design of a new nanosystem that allows for the control of Dox loading and delivery results is a powerful tool to control Dox release only in cancer cells. For this reason, supramolecular self-assembly was performed between a poly(amidoamine) (PAMAM) dendrimer decorated with four β-cyclodextrin (βCD) units (PAMAM-βCD) and an adamantane-hydrazone-doxorubicin (Ad-h-Dox) prodrug. Methods: The formation of inclusion complexes (ICs) between the prodrug and all the βCD cavities present on the surface of the PAMAM-βCD dendrimer was followed by 1H-NMR titration and corroborated by 2D NOESY experiments. A full characterization of the supramolecular assembly was performed in the solid state by thermal analysis (DSC/TGA) and scanning electron microscopy (SEM) and in solution by the DOSY NMR technique in D2O. Furthermore, the Dox release profiles from the PAMAM-βCD/Ad-h-Dox assembly at different pH values was studied by comparing the efficiency against a native βCD/Ad-h-Dox IC. Additionally, in vitro cytotoxic activity assays were performed for the nanocarrier alone and the two supramolecular assemblies in different carcinogenic cell lines. Results: The PAMAM-βCD/Ad-h-Dox assembly was adequately characterized, and the cytotoxic activity results demonstrate that the nanocarrier alone and its hydrolysis product are innocuous compared to the PAMAM-βCD/Ad-h-Dox nanocarrier that showed cytotoxicity equivalent to free Dox in the tested cancer cell lines. The in vitro drug release assays for the PAMAM-βCD/Ad-h-Dox system showed an acidic pH-dependent behavior and a prolonged profile of up to more than 72 h. Conclusions: The design of PAMAM-βCD/Ad-h-Dox consists of a new controlled and prolonged Dox release system for potential use in cancer treatment.
Collapse
Affiliation(s)
- Kendra Sorroza-Martínez
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, Calzada del Hueso 1100, Col. Villa Quietud, Mexico City CP 04960, Mexico;
| | - Ignacio González-Sánchez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Circuito Escolar, Ciudad Universitaria, Mexico City CP 04510, Mexico; (I.G.-S.); (M.C.)
| | - Raúl Villamil-Ramos
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca CP 62209, Mexico; (R.V.-R.); (J.A.G.-Á.)
| | - Marco Cerbón
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Circuito Escolar, Ciudad Universitaria, Mexico City CP 04510, Mexico; (I.G.-S.); (M.C.)
| | - Jorge Antonio Guerrero-Álvarez
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca CP 62209, Mexico; (R.V.-R.); (J.A.G.-Á.)
| | - Cristina Coronel-Cruz
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Circuito Escolar, Ciudad Universitaria, Mexico City CP 04510, Mexico;
| | - Ernesto Rivera
- Departamento de Reología, Instituto de Investigaciones en Materiales, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Mexico City CP 04510, Mexico
| | - Israel González-Méndez
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001, Col. Chamilpa, Cuernavaca CP 62209, Mexico; (R.V.-R.); (J.A.G.-Á.)
| |
Collapse
|
44
|
shaikh R, Bhattacharya S, Saoji SD. Development, optimization, and characterization of polymeric micelles to improve dasatinib oral bioavailability: Hep G2 cell cytotoxicity and in vivo pharmacokinetics for targeted liver cancer therapy. Heliyon 2024; 10:e39632. [PMID: 39559212 PMCID: PMC11570312 DOI: 10.1016/j.heliyon.2024.e39632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/20/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
The efficacy of dasatinib (DAS) in treating hepatocellular carcinoma (HCC) is hindered by its poor bioavailability, limiting its clinical potential. In this study, we explored the use of TPGS-Soluplus micelles as an innovative drug delivery platform to enhance DAS solubility, stability, and therapeutic impact. A series of TPGS-Soluplus copolymers were synthesized, varying the D-α-tocopheryl polyethylene glycol succinate (TPGS) forms (1000, 2000, and 3500) and adjusting the TPGS to Soluplus weight ratios (1:1, 1:2, and 1:3). Our goal was to identify the optimal formulation with the highest entrapment efficiency, smallest particle size, and enhanced drug loading. The TPGS1000-Soluplus copolymer, with a DAS-to-polymer ratio of 1:30 and a TPGS ratio of 1:2, demonstrated superior performance, achieving an entrapment efficiency of 64.479 ± 1.45 % and drug loading of 5.05 ± 1.01 %. The DAS-loaded micelles (DAS-PMs) exhibited a notably small particle size of 64.479 ± 1.45 nm and demonstrated controlled release kinetics, with 85.60 ± 5.4 % of the drug released over 72 h. Cellular uptake studies using Hep G2 cells revealed significantly enhanced absorption of DAS-PMs compared to free DAS, reflected in lower IC50 values in MTT assays at 24 and 48 h. Pharmacokinetic analysis further highlighted the benefits of the DAS-PMs, with an AUC0-∞ 2.16 times higher and mean residual time (MRT) 1.3 times longer than free DAS, a statistically significant improvement (p < 0.01). These findings suggest that TPGS-Soluplus micelles offer a promising strategy for improving the bioavailability and efficacy of DAS in HCC treatment, presenting a potential new therapeutic avenue for patients with limited options. This innovative formulation could significantly enhance DAS delivery, potentially leading to improved clinical outcomes in liver cancer therapy.
Collapse
Affiliation(s)
- Rehan shaikh
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM’S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
| | - Suprit D. Saoji
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University Nagpur, Mahatma Jyotiba Fuley Shaikshanik Parisar, University Campus, Amravati Road, Nagpur, 440033, Maharashtra, India
| |
Collapse
|
45
|
Haghighi E, Abolmaali SS, Dehshahri A, Mousavi Shaegh SA, Azarpira N, Tamaddon AM. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. J Nanobiotechnology 2024; 22:710. [PMID: 39543630 PMCID: PMC11566655 DOI: 10.1186/s12951-024-02972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
RNA therapeutics, such as mRNA, siRNA, and CRISPR-Cas9, present exciting avenues for treating diverse diseases. However, their potential is commonly hindered by vulnerability to degradation and poor cellular uptake, requiring effective delivery systems. Lipid nanoparticles (LNPs) have emerged as a leading choice for in vivo RNA delivery, offering protection against degradation, enhanced cellular uptake, and facilitation of endosomal escape. However, LNPs encounter numerous challenges for targeted RNA delivery in vivo, demanding advanced particle engineering, surface functionalization with targeting ligands, and a profound comprehension of the biological milieu in which they function. This review explores the structural and physicochemical characteristics of LNPs, in-vivo fate, and customization for RNA therapeutics. We highlight the quality-by-design (QbD) approach for targeted delivery beyond the liver, focusing on biodistribution, immunogenicity, and toxicity. In addition, we explored the current challenges and strategies associated with LNPs for in-vivo RNA delivery, such as ensuring repeated-dose efficacy, safety, and tissue-specific gene delivery. Furthermore, we provide insights into the current clinical applications in various classes of diseases and finally prospects of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
- Elahe Haghighi
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Azarpira
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
46
|
Geng S, Fang B, Wang K, Wang F, Zhou Y, Hou Y, Iqbal MZ, Chen Y, Yu Z. Polydopamine Nanoformulations Induced ICD and M1 Phenotype Macrophage Polarization for Enhanced TNBC Synergistic Photothermal Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59814-59832. [PMID: 39450881 DOI: 10.1021/acsami.4c11594] [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: 10/26/2024]
Abstract
Photothermal therapy (PTT) is a promising technology that can achieve the thermal ablation of tumors and induce immunogenic cell death (ICD). However, relying solely on the antitumor immune responses caused by PTT-induced ICD is insufficient to suppress tumor metastasis and recurrence effectively. Fortunately, multifunctional nanoformulation-based synergistic photothermal immunotherapy can eliminate primary and metastatic tumors and inhibit tumor recurrence for a long time. Herein, we select polydopamine (PDA) nanoparticles to serve as the carrier for our nanomedicine as well as a potent photothermal agent and modulator of macrophage polarization. PDA nanoparticles are loaded with the insoluble immune adjuvant Imiquimod (R837) to construct PDA(R837) nanoformulations. These straightforward yet highly effective nanoformulations demonstrate excellent performance, allowing for successful triple-negative breast cancer (TNBC) treatment through synergistic photothermal immunotherapy. Moreover, experimental results showed that PDA(R837) implementation of PTT is effective in the thermal ablation of primary tumors while causing ICD and releasing R837, further promoting dendritic cell (DC) maturation and activating the systemic antitumor immune response. Furthermore, PDA(R837) nanoformulations inhibit tumor metastasis and recurrence and achieve M1 phenotype macrophage polarization, achieving long-term and excellent antitumor efficacy. Therefore, the structurally simple PDA(R837) nanoformulations provide cancer treatment and have excellent clinical translational application prospects.
Collapse
Affiliation(s)
- Siqi Geng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Baoru Fang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Ke Wang
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Fang Wang
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P. R. China
| | - Yiqing Zhou
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Yike Hou
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P. R. China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P. R. China
| | - Yanping Chen
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| | - Zhangsen Yu
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
- Laboratory of Nanomedicine, Medical Science Research Center, School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, P. R. China
| |
Collapse
|
47
|
Hulugalla K, Shofolawe-Bakare O, Toragall VB, Mohammad SA, Mayatt R, Hand K, Anderson J, Chism C, Misra SK, Shaikh T, Tanner EEL, Smith AE, Sharp JS, Fitzkee NC, Werfel T. Glycopolymeric Nanoparticles Enrich Less Immunogenic Protein Coronas, Reduce Mononuclear Phagocyte Clearance, and Improve Tumor Delivery Compared to PEGylated Nanoparticles. ACS NANO 2024; 18:30540-30560. [PMID: 39436672 DOI: 10.1021/acsnano.4c08922] [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] [Indexed: 10/23/2024]
Abstract
Nanoparticles (NPs) offer significant promise as drug delivery vehicles; however, their in vivo efficacy is often hindered by the formation of a protein corona (PC), which influences key physiological responses such as blood circulation time, biodistribution, cellular uptake, and intracellular localization. Understanding NP-PC interactions is crucial for optimizing NP design for biomedical applications. Traditional approaches have utilized hydrophilic polymer coatings like polyethylene glycol (PEG) to resist protein adsorption, but glycopolymer-coated nanoparticles have emerged as potential alternatives due to their biocompatibility and ability to reduce the adsorption of highly immunogenic proteins. In this study, we synthesized and characterized glycopolymer-based poly[2-(diisopropylamino)ethyl methacrylate-b-poly(methacrylamidoglucopyranose) (PDPA-b-PMAG) NPs as an alternative to PEGylated NPs. We characterized the polymers using a range of techniques to establish their molecular weight and chemical composition. PMAG and PEG-based NPs showed equivalent physicochemical properties with sizes of ∼100 nm, spherical morphology, and neutral surface charges. We next assessed the magnitude of protein adsorption on both NPs and catalogued the identity of the adsorbed proteins using mass spectrometry-based techniques. The PMAG NPs were found to adsorb fewer proteins in vitro as well as fewer immunogenic proteins such as Immunoglobulins and Complement proteins. Flow cytometry and confocal microscopy were employed to examine cellular uptake in RAW 264.7 macrophages and MDA-MB-231 tumor cells, where PMAG NPs showed higher uptake into tumor cells over macrophages. In vivo studies in BALB/c mice with orthotopic 4T1 breast cancer xenografts showed that PMAG NPs exhibited prolonged circulation times and enhanced tumor accumulation compared to PEGylated NPs. The biodistribution analysis also revealed greater selectivity for tumor tissue over the liver for PMAG NPs. These findings highlight the potential of glycopolymeric NPs to improve tumor targeting and reduce macrophage uptake compared to PEGylated NPs, offering significant advancements in cancer nanomedicine and immunotherapy.
Collapse
Affiliation(s)
- Kenneth Hulugalla
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Oluwaseyi Shofolawe-Bakare
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Veeresh B Toragall
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Sk Arif Mohammad
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Railey Mayatt
- Department of Chemistry, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Kelsie Hand
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Joshua Anderson
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Claylee Chism
- Department of Chemistry and Biochemistry, University of Mississippi, University, Mississippi 38677, United States
| | - Sandeep K Misra
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Tanveer Shaikh
- Department of Chemistry, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Eden E L Tanner
- Department of Chemistry and Biochemistry, University of Mississippi, University, Mississippi 38677, United States
| | - Adam E Smith
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
| | - Joshua S Sharp
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
| | - Nicholas C Fitzkee
- Department of Chemistry, Mississippi State University, Starkville, Mississippi 39762, United States
| | - Thomas Werfel
- Department of BioMolecular Sciences, University of Mississippi, University, Mississippi 38677, United States
- Department of Biomedical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Department of Chemical Engineering, University of Mississippi, University, Mississippi 38677, United States
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, Mississippi 39216, United States
| |
Collapse
|
48
|
Abo Qoura L, Morozova E, Ramaa СS, Pokrovsky VS. Smart nanocarriers for enzyme-activated prodrug therapy. J Drug Target 2024; 32:1029-1051. [PMID: 39045650 DOI: 10.1080/1061186x.2024.2383688] [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: 03/18/2024] [Revised: 06/26/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Exogenous enzyme-activated prodrug therapy (EPT) is a potential cancer treatment strategy that delivers non-human enzymes into or on the surface of the cell and subsequently converts a non-toxic prodrug into an active cytotoxic substance at a specific location and time. The development of several pharmacological pairs based on EPT has been the focus of anticancer research for more than three decades. Numerous of these pharmacological pairs have progressed to clinical trials, and a few have achieved application in specific cancer therapies. The current review highlights the potential of enzyme-activated prodrug therapy as a promising anticancer treatment. Different microbial, plant, or viral enzymes and their corresponding prodrugs that advanced to clinical trials have been listed. Additionally, we discuss new trends in the field of enzyme-activated prodrug nanocarriers, including nanobubbles combined with ultrasound (NB/US), mesoscopic-sized polyion complex vesicles (PICsomes), nanoparticles, and extracellular vesicles (EVs), with special emphasis on smart stimuli-triggered drug release, hybrid nanocarriers, and the main application of nanotechnology in improving prodrugs.
Collapse
Affiliation(s)
- Louay Abo Qoura
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), Moscow, Russia
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Elena Morozova
- Engelhardt Institute of Molecular Biology of the, Russian Academy of Sciences, Moscow, Russia
| | - С S Ramaa
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Mumbai, India
| | - Vadim S Pokrovsky
- Research Institute of Molecular and Cellular Medicine, People's Friendship University of Russia (RUDN University), Moscow, Russia
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
49
|
Lorenc P, Sikorska A, Molenda S, Guzniczak N, Dams-Kozlowska H, Florczak A. Physiological and tumor-associated angiogenesis: Key factors and therapy targeting VEGF/VEGFR pathway. Biomed Pharmacother 2024; 180:117585. [PMID: 39442237 DOI: 10.1016/j.biopha.2024.117585] [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: 08/01/2024] [Revised: 10/03/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Cancer remains one of the leading causes of death worldwide and poses a significant challenge to effective treatment due to its complexity. Angiogenesis, the formation of new blood vessels, is one of the cancer hallmarks and is a critical process in tumor growth and metastasis. The pivotal role of angiogenesis in cancer development has made antiangiogenic treatment a promising strategy for cancer therapy. To develop an effective therapy, it is essential to understand the basics of the physiological and tumor angiogenesis process. This review presents the primary factors related to physiological and tumor angiogenesis and the mechanisms of angiogenesis in tumors. We summarize potential molecular targets for cancer treatment by focusing on the vasculature, with the VEGF/VEGFR pathway being one of the most important and well-studied. Additionally, we present the advantages and limitations of currently used clinical protocols for cancer treatment targeting the VEGF/VEGFR pathway.
Collapse
Affiliation(s)
- Patryk Lorenc
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland; Doctoral School, Poznan University of Medical Sciences, 70 Bukowska St, Poznan 60-812, Poland
| | - Agata Sikorska
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland
| | - Sara Molenda
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland; Doctoral School, Poznan University of Medical Sciences, 70 Bukowska St, Poznan 60-812, Poland
| | - Natalia Guzniczak
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland
| | - Anna Florczak
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61‑866, Poland.
| |
Collapse
|
50
|
Lee LC, Lo KK. Leveraging the Photofunctions of Transition Metal Complexes for the Design of Innovative Phototherapeutics. SMALL METHODS 2024; 8:e2400563. [PMID: 39319499 PMCID: PMC11579581 DOI: 10.1002/smtd.202400563] [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: 04/19/2024] [Revised: 08/03/2024] [Indexed: 09/26/2024]
Abstract
Despite the advent of various medical interventions for cancer treatment, the disease continues to pose a formidable global health challenge, necessitating the development of new therapeutic approaches for more effective treatment outcomes. Photodynamic therapy (PDT), which utilizes light to activate a photosensitizer to produce cytotoxic reactive oxygen species (ROS) for eradicating cancer cells, has emerged as a promising approach for cancer treatment due to its high spatiotemporal precision and minimal invasiveness. However, the widespread clinical use of PDT faces several challenges, including the inefficient production of ROS in the hypoxic tumor microenvironment, the limited penetration depth of light in biological tissues, and the inadequate accumulation of photosensitizers at the tumor site. Over the past decade, there has been increasing interest in the utilization of photofunctional transition metal complexes as photosensitizers for PDT applications due to their intriguing photophysical and photochemical properties. This review provides an overview of the current design strategies used in the development of transition metal complexes as innovative phototherapeutics, aiming to address the limitations associated with PDT and achieve more effective treatment outcomes. The current challenges and future perspectives on the clinical translation of transition metal complexes are also discussed.
Collapse
Affiliation(s)
- Lawrence Cho‐Cheung Lee
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| | - Kenneth Kam‐Wing Lo
- Department of ChemistryCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
- State Key Laboratory of Terahertz and Millimeter WavesCity University of Hong KongTat Chee AvenueKowloonHong KongP. R. China
| |
Collapse
|