1
|
Rajana N, Naraharisetti LT, Kumari NU, Vasave R, Chigurupati SPD, Sharma A, Godugu C, Mehra NK. Fabrication and characterization of teriflunomide-loaded chondroitin sulfate hybridized zein nanoparticles for the management of triple negative breast cancer. Int J Biol Macromol 2025; 300:140316. [PMID: 39864685 DOI: 10.1016/j.ijbiomac.2025.140316] [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: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 01/28/2025]
Abstract
The objective of this work was to explore the Teriflunomide (TFM) -loaded chondroitin sulfate hybridized zein nanoparticles (TZCNPs) for the treatment of triple-negative breast cancer (TNBC). The particle size, PDI and %EE of optimized TZCNPs was found 208.7 ± 7.26 nm,0.173 ± 0.004, and 80.18 ± 1.03 %, respectively. TZCNPs demonstrate a 7-10 folds increase in cytotoxicity against free TFM in MDA-MB-231 cells and a 4-6 folds increase in MCF-7 cells, respectively. CD44 receptor blocking resulted in a 3.4-fold reduction in anti-cancer efficacy and a 1.7-fold decrease in cellular uptake of TZCNPs in MDA-MB-231 cells, significantly/strongly indicating the critical role of the CD44-mediated uptake mechanism. TZCNPs displayed enhanced apoptosis, mitochondrial depolarization, ROS generation, cell invasion inhibition, and inhibited colony formation compared to free TFM in MDA-MB-231 cells. TZCNPs exhibited approximately 6.8-fold enhanced cytotoxicity and a 1.66-fold decrease in spheroid volume in a multicellular tumor spheroid model of MDA-MB-231 cells compared to free TFM. TZCNPs also exhibited greater disintegration of spheroids and more dead cells (live/dead staining). In pharmacokinetic studies, TZCNPs displayed reduced CL and enhanced the AUC, MRT, and t ½ by 3.64-fold, 2.17-fold, 1.83-fold, and 1.73-fold than the free TFM suspension. An acute toxicity study revealed a good safety profile of TZCNPs, which could be a potential treatment option for TNBC.
Collapse
Affiliation(s)
- Naveen Rajana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Lakshmi Tulasi Naraharisetti
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Nalla Usha Kumari
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Ravindra Vasave
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Sri Pada Datta Chigurupati
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India.
| |
Collapse
|
2
|
Argitekin E, Erez O, Cakan-Akdogan G, Akdogan Y. Periodate-Mediated Cross-Linking for the Preparation of Catechol Conjugated Albumin Nanoparticles Used for in Vitro Drug Delivery. ACS APPLIED BIO MATERIALS 2025; 8:2182-2193. [PMID: 39950848 PMCID: PMC11931531 DOI: 10.1021/acsabm.4c01737] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/01/2025] [Accepted: 02/02/2025] [Indexed: 03/18/2025]
Abstract
Conjugation of serum albumin protein with catechol-containing dopamine molecules provides an alternative method for the preparation of albumin nanoparticles (NPs). A commonly used desolvation method utilizes glutaraldehyde as a cross-linking agent. Here, the catechol cross-linking mechanism is used instead of glutaraldehyde providing advantages to prevent toxicity and an undesirable reaction of glutaraldehyde with cargo molecules. Covalent cross-linking between dopamine conjugated bovine serum albumin (D-BSA) proteins was obtained in the presence of sodium periodate (NaIO4) as an oxidizer. As a result, spherical D-BSA NPs with a uniform size distribution of around 100 nm in diameter and negative zeta potential around -28 mV were prepared. Optimal conditions were reached when a dopamine:IO4- molar ratio of 2:1, pH 7.4 of the medium, and acetone as the desolvating agent were used. Furthermore, the obtained NPs display antioxidant properties, have rapid biodegradability in the presence of trypsin, and have a high doxorubicin (DOX) loading (9.1%) with a sustainable drug release. DOX loaded D-BSA NPs also caused up to 90% breast cancer cell (MCF-7) death within 24 h. These results show that drug carrying albumin NPs can alternatively be prepared via covalently cross-linked catechol groups and used in drug delivery studies.
Collapse
Affiliation(s)
- Eda Argitekin
- Materials
Science and Engineering Department, Izmir
Institute of Technology, Izmir 35430, Turkey
| | - Ozlem Erez
- Izmir
Biomedicine and Genome Center, 35340 Izmir, Turkey
| | - Gulcin Cakan-Akdogan
- Izmir
Biomedicine and Genome Center, 35340 Izmir, Turkey
- Izmir
International Biomedicine and Genome Institute, Dokuz Eylul University, 35430 Izmir, Turkey
| | - Yasar Akdogan
- Materials
Science and Engineering Department, Izmir
Institute of Technology, Izmir 35430, Turkey
| |
Collapse
|
3
|
Wang K, Shang J, Tao C, Huang M, Wei D, Yang L, Yang J, Fan Q, Ding Q, Zhou M. Advancements in Betulinic Acid-Loaded Nanoformulations for Enhanced Anti-Tumor Therapy. Int J Nanomedicine 2024; 19:14075-14103. [PMID: 39748899 PMCID: PMC11694648 DOI: 10.2147/ijn.s493489] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
Betulinic acid (BA) is a natural compound obtained from plant extracts and is known for its diverse pharmacological effects, including anti-tumor, antibacterial, anti-inflammatory, antiviral, and anti-atherosclerotic properties. Its potential in anti-tumor therapy has garnered considerable attention, particularly for the treatment of breast, lung, and liver cancers. However, the clinical utility of BA is greatly hindered by its poor water solubility, low bioavailability, and off-target toxicity. To address these issues, researchers have developed various BA-loaded nanoformulations, such as nanoparticles, liposomes, micelles, and nanofibers, aiming to improve its solubility and bioavailability, prolong plasma half-life, and enhance targeting ability, thereby augmenting its anti-cancer efficacy. In preparing this review, we conducted extensive searches in well-known databases, including PubMed, Web of Science, and ScienceDirect, using keywords like "betulinic acid", "nanoparticles", "drug delivery", "tumor", and "cancer", covering the literature from 2014 to 2024. The review provides a comprehensive overview of recent advancements in the application of BA-loaded nano-delivery systems for anti-tumor therapy and offers insights into their future development prospects.
Collapse
Affiliation(s)
- Ke Wang
- Department of Pharmacy, the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jinlu Shang
- Department of Pharmacy, West China Hospital Sichuan University Jintang Hospital, Chengdu, People’s Republic of China
| | - Chao Tao
- Department of Pharmacy, the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
- Department of Clinical Pharmacy, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
| | - Mingquan Huang
- Sichuan Treatment Center for Gynaecologic and Breast Diseases (Breast Surgery), the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Daiqing Wei
- Department of Orthopaedics, the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Liuxuan Yang
- Department of Pharmacy, the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jing Yang
- Department of Pharmacy, the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Qingze Fan
- Department of Pharmacy, the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| | - Qian Ding
- Department of Clinical Pharmacy, the Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, People’s Republic of China
| | - Meiling Zhou
- Department of Pharmacy, the Affiliated Hospital, Southwest Medical University, Luzhou, People’s Republic of China
| |
Collapse
|
4
|
Vijayakumar S, González-Sánchez ZI, Divya M, Amanullah M, Durán-Lara EF, Li M. Efficacy of chondroitin sulfate as an emerging biomaterial for cancer-targeted drug delivery: A short review. Int J Biol Macromol 2024; 283:137704. [PMID: 39549800 DOI: 10.1016/j.ijbiomac.2024.137704] [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: 08/25/2024] [Revised: 11/11/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
The global increase in cancer incidence over the past decade highlights the urgent need for more effective therapeutic strategies. Conventional cancer treatments face challenges such as drug resistance and off-target toxicity, which affect healthy tissues. Chondroitin sulfate (CHDS), a naturally occurring bioactive macromolecule, has gained attention because of its biocompatibility, biodegradability, and low toxicity, positioning it as an ideal candidate for cancer-targeted drug delivery systems. This review highlights the potential of CHDS as an emerging biomaterial in cancer therapy, focusing on its unique biological properties and applications in drug delivery platforms. Furthermore, we discuss the advantages of CHDS-based biomaterials in enhancing cancer treatment efficacy and minimizing side effects, in order to provide a comprehensive reference for future research on CHDS-based cancer therapeutics.
Collapse
Affiliation(s)
- Sekar Vijayakumar
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| | - Zaira I González-Sánchez
- Nanobiology Laboratory, Department of Natural and Exact Sciences, Pontificia Universidad Católica Madre y Maestra, PUCMM, Autopista Duarte Km 1 ½, Santiago de los Caballeros, Dominican Republic; Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Mani Divya
- Advanced Laboratory of Bio-nanomaterials, BioMe Live Analytical Centre, Kannappa Tower, College Road, Karaikudi - 630 003, Tamilnadu, India
| | - Mohammed Amanullah
- Department of clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Esteban F Durán-Lara
- Bio&NanoMaterialsLab Drug Delivery and Controlled Release, Universidad de Talca, Talca 3460000, Maule, Chile; Departamento de Microbiología, Facultad de Ciencias de la Salud, Universidad de Talca, Talca 3460000, Maule, Chile
| | - Mingchun Li
- College of Material Science and Engineering, Huaqiao University, Engineering Research Center of Environment-Friendly Functional Materials, Ministry of Education, Xiamen 361021, PR China.
| |
Collapse
|
5
|
Gunjkar S, Gupta U, Nair R, Paul P, Aalhate M, Mahajan S, Maji I, Chourasia MK, Guru SK, Singh PK. The Neoteric Paradigm of Biomolecule-Functionalized Albumin-Based Targeted Cancer Therapeutics. AAPS PharmSciTech 2024; 25:265. [PMID: 39500822 DOI: 10.1208/s12249-024-02977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/17/2024] [Indexed: 12/12/2024] Open
Abstract
Albumin is a nature-derived, versatile protein carrier, that has been explored extensively by researchers for anticancer drug delivery due to its role in enhancing drug stability, solubility, circulation time, targeting capabilities, and overall therapeutic efficacy. Albumin nanoparticles possess inherent biocompatibility, biodegradability, and passive tumor-targeting ability due to the enhanced permeability and retention effect. However, non-specific accumulation of cytotoxic agents in healthy tissues remains a challenge. In this paper, the functionalization of albumin nanoparticles using various biomolecules including antibodies, nucleic acids, proteins and peptides, vitamins, chondroitin sulfate, hyaluronic acid, and lactobionic acid have been discussed which enables specific recognition and binding to cancer cells. Furthermore, we highlight the supremacy of such a targeted approach in tumor-specific drug delivery, minimization of off-target effects, potential improvement in therapeutic efficacy, cellular internalization, reduced side effects, and better clinical outcomes. This review centers on how they have revolutionized the field of biomedical research and tuned into an excellent targeted approach. In conclusion, this review highlights in detail the role of albumin as a nanocarrier for tumor-targeted delivery using biomolecules as ligands.
Collapse
Affiliation(s)
- Swati Gunjkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Rahul Nair
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Priti Paul
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Indrani Maji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Manish K Chourasia
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, U.P., India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, Telangana, India.
| |
Collapse
|
6
|
Song Q, Wang P, Wu J, Lu M, Xia Q, Shi Y, Wang Z, Ma X, Zhao Q. Analysis of the role of CHPF in colorectal cancer tumorigenesis and immunotherapy based on bioinformatics and experiments. Discov Oncol 2024; 15:458. [PMID: 39292317 PMCID: PMC11410747 DOI: 10.1007/s12672-024-01340-2] [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: 07/10/2023] [Accepted: 09/11/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Chondroitin polymerizing factor (CHPF) has been found to be involved in the development of numerous cancers and correlated with poor prognosis. However, its role in the tumorigenesis and development of colorectal cancer (CRC) remains unknown. METHODS In our research, we explored CHPF expression and clinicopathological characteristics using The Cancer Genome Atlas Program (TCGA), UALCAN, GSE9348, TIMER2.0 and The Human Protein Atlas (HPA) database, in addition, we validated CHPF expression in CRC cell lines by Real-Time Quantitative PCR (qRT-PCR) and Western blot (WB). KM-Plotter, PrognoScan and TCGA were also utilized to verify its prognosis value in CRC. Small-interfer RNA (Si-RNA) was used to perform Cell Counting Kit-8 (CCK8), colony formation, 5-ethynyl-2'-deoxyuridine (EDU), transwell and wound healing assays to testify its function on the tumor progression. Based on TCGA database, we probed potential biological mechanism by which CHPF play its role via clusterProfiler package and GEPIA database and we validated their correlation by WB assay. Moreover, we explored its potential association with the tumor microenvironment (TME), immune infiltrated cells, immune checkpoints, tumor mutation burden (TMB) as well as microsatellite instability (MSI), and investigated immunotherapy sensitivity via Tumor Immune Dysfunction and Exclusion (TIDE) algorithm as well as potentially effective therapeutic drugs via pRRophetic algorithm. RESULTS CHPF was identified upregulated in CRC tissues and cells, correlated with poor prognosis, and nodal metastasis status, stage and histological subtype. Down-regulation of CHPF inhibited CRC cell proliferation, migration and its expression correlated with wnt pathway key molecules. In addition, high expression of CHPF was positively correlated with TME scores, Regulatory T cells (Tregs) cell infiltration degree, Programmed death-1 (PD-1), MSI-high (MSI-H), and TIDE scores, however, not with TMB. Targeted drug analysis showed that patients with high CHPF expression were more sensitive to telatinib, recaparib, serdemetan, and trametinib. CONCLUSION CHPF could promote the proliferation and migration of CRC cells and lead to poor prognosis, possibly through wnt pathways as well as changes in TME. Patients with high expression of CHPF had poor efficacy in immunotherapy, which might be related to Tregs cell infiltration. Above all, it might offer more reliable guidance for future immunotherapy.
Collapse
Affiliation(s)
- Qingyu Song
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pengchao Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingyu Wu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qingcheng Xia
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yexin Shi
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zijun Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Ma
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Qinghong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
7
|
Dhiman R, Bazad N, Mukherjee R, Himanshu, Gunjan, Leal E, Ahmad S, Kaur K, Raj VS, Chang CM, Pandey RP. Enhanced drug delivery with nanocarriers: a comprehensive review of recent advances in breast cancer detection and treatment. DISCOVER NANO 2024; 19:143. [PMID: 39243326 PMCID: PMC11380656 DOI: 10.1186/s11671-024-04086-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024]
Abstract
Breast cancer (BC) remains a leading cause of morbidity and mortality among women worldwide, with triple-negative breast cancer (TNBC) posing significant treatment challenges due to its aggressive phenotype and resistance to conventional therapies. Recent advancements in nanocarrier technology offer promising solutions for enhancing drug delivery, improving bioavailability, and increasing drug accumulation at tumor sites through targeted approaches. This review delves into the latest innovations in BC detection and treatment, highlighting the role of nanocarriers like polymeric micelles, liposomes, and magnetic nanoparticles in overcoming the limitations of traditional therapies. Additionally, the manuscript discusses the integration of cutting-edge diagnostic tools, such as multiplex PCR-Nested Next-Generation Sequencing (mPCR-NGS) and blood-based biomarkers, which are revolutionizing early detection and molecular profiling of BC. The convergence of these technologies not only enhances therapeutic outcomes but also paves the way for personalized medicine in BC management. This comprehensive review underscores the potential of nanocarriers in transforming BC treatment and emphasizes the critical importance of early detection in improving patient prognosis.
Collapse
Affiliation(s)
- Ruby Dhiman
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
- School of Health Sciences and Technology (SOHST), UPES, Dehradun, Uttarakhand, 248007, India
| | - Nancy Bazad
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - Riya Mukherjee
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Himanshu
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Gunjan
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC
| | - Elcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belem, Pará, Brazil
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Hail City, Kingdom of Saudi Arabia
| | - Kirtanjot Kaur
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - V Samuel Raj
- Centre for Drug Design Discovery and Development (C4D), SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - Chung-Ming Chang
- Department in Biotechnology Industry, Chang Gung University, Taoyuan City, Taiwan, ROC.
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan City, Taiwan, ROC.
- Laboratory Animal Center, Chang Gung University, Taoyuan City, Taiwan, ROC.
| | - Ramendra Pati Pandey
- School of Health Sciences and Technology (SOHST), UPES, Dehradun, Uttarakhand, 248007, India.
| |
Collapse
|
8
|
Cao Y, Xu R, Liang Y, Tan J, Guo X, Fang J, Wang S, Xu L. Nature-inspired protein mineralization strategies for nanoparticle construction: advancing effective cancer therapy. NANOSCALE 2024; 16:13718-13754. [PMID: 38954406 DOI: 10.1039/d4nr01536c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Recently, nanotechnology has shown great potential in the field of cancer therapy due to its ability to improve the stability and solubility and reduce side effects of drugs. The biomimetic mineralization strategy based on natural proteins and metal ions provides an innovative approach for the synthesis of nanoparticles. This strategy utilizes the unique properties of natural proteins and the mineralization ability of metal ions to combine nanoparticles through biomimetic mineralization processes, achieving the effective treatment of tumors. The precise control of the mineralization process between proteins and metal ions makes it possible to obtain nanoparticles with the ideal size, shape, and surface characteristics, thereby enhancing their stability and targeting ability in vivo. Herein, initially, we analyze the role of protein molecules in biomineralization and comprehensively review the functions, properties, and applications of various common proteins and metal particles. Subsequently, we systematically review and summarize the application directions of nanoparticles synthesized based on protein biomineralization in tumor treatment. Specifically, we discuss their use as efficient drug delivery carriers and role in mediating monotherapy and synergistic therapy using multiple modes. Also, we specifically review the application of nanomedicine constructed through biomimetic mineralization strategies using natural proteins and metal ions in improving the efficiency of tumor immunotherapy.
Collapse
Affiliation(s)
- Yuan Cao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Rui Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Yixia Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Jiabao Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Xiaotang Guo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Junyue Fang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| | - Shibo Wang
- Institute of Smart Biomaterials, School of Materials Science and Engineering and Zhejiang Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, P. R. China
| | - Lei Xu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China.
- Guangzhou Key Laboratory of Medical Nanomaterials, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, P. R. China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-Sen Memorial Hospital, Foshan 528200, P. R. China
| |
Collapse
|
9
|
Xu Y, Bai Z, Lan T, Fu C, Cheng P. CD44 and its implication in neoplastic diseases. MedComm (Beijing) 2024; 5:e554. [PMID: 38783892 PMCID: PMC11112461 DOI: 10.1002/mco2.554] [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: 08/01/2023] [Revised: 03/20/2024] [Accepted: 04/01/2024] [Indexed: 05/25/2024] Open
Abstract
CD44, a nonkinase single span transmembrane glycoprotein, is a major cell surface receptor for many other extracellular matrix components as well as classic markers of cancer stem cells and immune cells. Through alternative splicing of CD44 gene, CD44 is divided into two isoforms, the standard isoform of CD44 (CD44s) and the variant isoform of CD44 (CD44v). Different isoforms of CD44 participate in regulating various signaling pathways, modulating cancer proliferation, invasion, metastasis, and drug resistance, with its aberrant expression and dysregulation contributing to tumor initiation and progression. However, CD44s and CD44v play overlapping or contradictory roles in tumor initiation and progression, which is not fully understood. Herein, we discuss the present understanding of the functional and structural roles of CD44 in the pathogenic mechanism of multiple cancers. The regulation functions of CD44 in cancers-associated signaling pathways is summarized. Moreover, we provide an overview of the anticancer therapeutic strategies that targeting CD44 and preclinical and clinical trials evaluating the pharmacokinetics, efficacy, and drug-related toxicity about CD44-targeted therapies. This review provides up-to-date information about the roles of CD44 in neoplastic diseases, which may open new perspectives in the field of cancer treatment through targeting CD44.
Collapse
Affiliation(s)
- Yiming Xu
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ziyi Bai
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Tianxia Lan
- Department of BiotherapyLaboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Chenying Fu
- Laboratory of Aging and Geriatric Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Ping Cheng
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
10
|
Pang HL, Lu H, Liu P, Zhang YT, Zhang LT, Ren Q. A chondroitin sulfate purified from shark cartilage and bovine serum albumin interaction activity. Int J Biol Macromol 2024; 260:129499. [PMID: 38262829 DOI: 10.1016/j.ijbiomac.2024.129499] [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: 10/30/2023] [Revised: 12/31/2023] [Accepted: 01/12/2024] [Indexed: 01/25/2024]
Abstract
Chondroitin sulfate (CS) was extracted and purified from shark cartilage, and its interaction with bovine serum albumin (BSA) were studied. The content of chondroitin sulfate in shark cartilage was 29.97 % using the 1,9-dimethyl-methylene blue method. The molecular weight of CS was determined to be 62.464 kDa by high-performance gel permeation chromatography. UV and FT-IR spectroscopy identified the characteristics of CS and its functional group information. NMR spectroscopy and disaccharide derivatization revealed that CS was predominantly composed of disulfated disaccharides, specifically ΔDi4,6S. Fluorescence quenching experiments indicated that the interaction between CS and BSA exhibited static quenching, with a binding site number of 1. The binding process was primarily mediated by van der Waals forces and hydrogen bonds. Furthermore, synchronous and 3D fluorescence spectroscopy demonstrated that CS had minimal impact on the polarity and hydrophobicity of the microenvironment surrounding Tyr and Trp residues. UV-vis absorption and circular dichroism (CD) spectroscopy demonstrated the altered structure of BSA. The molecular docking analysis revealed that CS formed hydrogen bonds and salt bridges with BSA, predominantly binding to the IIA substructure domain of BSA. Investigating the interaction between CS and BSA holds the potential for enhancing its applications in drug delivery and tissue engineering endeavors.
Collapse
Affiliation(s)
- Hai-Long Pang
- Department of Pharmacy, Weifang Medical University, Weifang, Shandong, China; Department of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| | - Han Lu
- Department of Pharmacy, Weifang Medical University, Weifang, Shandong, China; Department of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| | - Peng Liu
- Rizhao Science and Technology Innovation Service Center, Rizhao, Shandong, China
| | - Yun-Tao Zhang
- Department of Pharmacy, Jining Medical University, Rizhao, Shandong, China.
| | - Li-Tao Zhang
- Department of Biological Science, Jining Medical University, Rizhao, Shandong, China.
| | - Qiang Ren
- Department of Pharmacy, Jining Medical University, Rizhao, Shandong, China.
| |
Collapse
|
11
|
Ji Q, Zhu H, Qin Y, Zhang R, Wang L, Zhang E, Zhou X, Meng R. GP60 and SPARC as albumin receptors: key targeted sites for the delivery of antitumor drugs. Front Pharmacol 2024; 15:1329636. [PMID: 38323081 PMCID: PMC10844528 DOI: 10.3389/fphar.2024.1329636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Albumin is derived from human or animal blood, and its ability to bind to a large number of endogenous or exogenous biomolecules makes it an ideal drug carrier. As a result, albumin-based drug delivery systems are increasingly being studied. With these in mind, detailed studies of the transport mechanism of albumin-based drug carriers are particularly important. As albumin receptors, glycoprotein 60 (GP60) and secreted protein acidic and rich in cysteine (SPARC) play a crucial role in the delivery of albumin-based drug carriers. GP60 is expressed on vascular endothelial cells and enables albumin to cross the vascular endothelial cell layer, and SPARC is overexpressed in many types of tumor cells, while it is minimally expressed in normal tissue cells. Thus, this review supplements existing articles by detailing the research history and specific biological functions of GP60 or SPARC and research advances in the delivery of antitumor drugs using albumin as a carrier. Meanwhile, the deficiencies and future perspectives in the study of the interaction of albumin with GP60 and SPARC are also pointed out.
Collapse
Affiliation(s)
- Qingzhi Ji
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
| | - Huimin Zhu
- Sheyang County Comprehensive Inspection and Testing Center, Yancheng, China
| | - Yuting Qin
- School of Pharmacy, Yancheng Teachers University, Yancheng, China
| | - Ruiya Zhang
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Lei Wang
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Erhao Zhang
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Xiaorong Zhou
- Department of Immunology, Medical School, Nantong University, Nantong, China
| | - Run Meng
- Department of Immunology, Medical School, Nantong University, Nantong, China
| |
Collapse
|
12
|
Hogan KJ, Perez MR, Mikos AG. Extracellular matrix component-derived nanoparticles for drug delivery and tissue engineering. J Control Release 2023; 360:888-912. [PMID: 37482344 DOI: 10.1016/j.jconrel.2023.07.034] [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: 03/16/2023] [Revised: 06/16/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
The extracellular matrix (ECM) consists of a complex combination of proteins, proteoglycans, and other biomolecules. ECM-based materials have been demonstrated to have high biocompatibility and bioactivity, which may be harnessed for drug delivery and tissue engineering applications. Herein, nanoparticles incorporating ECM-based materials and their applications in drug delivery and tissue engineering are reviewed. Proteins such as gelatin, collagen, and fibrin as well as glycosaminoglycans including hyaluronic acid, chondroitin sulfate, and heparin have been employed for cancer therapeutic delivery, gene delivery, and wound healing and regenerative medicine. Strategies for modifying and functionalizing these materials with synthetic and natural polymers or to enable stimuli-responsive degradation and drug release have increased the efficacy of these materials and nano-systems. The incorporation and modification of ECM-based materials may be used to drive drug targeting and increase tissue-specific cell differentiation more effectively.
Collapse
Affiliation(s)
- Katie J Hogan
- Department of Bioengineering, Rice University, Houston, TX, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA
| | - Marissa R Perez
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, USA.
| |
Collapse
|
13
|
Chen Z, Liu Z, Zhang Q, Huang S, Zhang Z, Feng X, Zeng L, Lin D, Wang L, Song H. Hypoxia-ameliorated photothermal manganese dioxide nanoplatform for reversing doxorubicin resistance. Front Pharmacol 2023; 14:1133011. [PMID: 36909187 PMCID: PMC9998484 DOI: 10.3389/fphar.2023.1133011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023] Open
Abstract
Drug resistance is a huge hurdle in tumor therapy. Tumor hypoxia contributes to chemotherapy resistance by inducing the hypoxia-inducible factor-1α (HIF-1α) pathway. To reduce tumor hypoxia, novel approaches have been devised, providing significant importance to reverse therapeutic resistance and improve the effectiveness of antitumor therapies. Herein, the nanosystem of bovine serum albumin (BSA)-templated manganese dioxide (MnO2) nanoparticles (BSA/MnO2 NPs) loaded with doxorubicin (DOX) (DOX-BSA/MnO2 NPs) developed in our previous report was further explored for their physicochemical properties and capacity to reverse DOX resistance because of their excellent photothermal and tumor microenvironment (TME) response effects. The DOX-BSA/MnO2 NPs showed good biocompatibility and hemocompatibility. Meanwhile, DOX-BSA/MnO2 NPs could greatly affect DOX pharmacokinetic properties, with prolonged circulation time and reduced cardiotoxicity, besides enhancing accumulation at tumor sites. DOX-BSA/MnO2 NPs can interact with H2O2 and H+ in TME to form oxygen and exhibit excellent photothermal effect to further alleviate hypoxia due to MnO2, reversing DOX resistance by down-regulating HIF-1α expression and significantly improving the antitumor efficiency in DOX-resistant human breast carcinoma cell line (MCF-7/ADR) tumor model. The hypoxia-ameliorated photothermal MnO2 platform is a promising strategy for revering DOX resistance.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Zhihong Liu
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Qian Zhang
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Sheng Huang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Zaizhong Zhang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Xianquan Feng
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Lingjun Zeng
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Ding Lin
- Department of Pharmacy, Jiaxing Maternal and Child Healthcare Hospital, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Lie Wang
- Department of General Surgery, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Hongtao Song
- Department of Pharmacy, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| |
Collapse
|
14
|
Lu R, Yu RJ, Yang C, Wang Q, Xuan Y, Wang Z, He Z, Xu Y, Kou L, Zhao YZ, Yao Q, Xu SH. Evaluation of the hepatoprotective effect of naringenin loaded nanoparticles against acetaminophen overdose toxicity. Drug Deliv 2022; 29:3256-3269. [PMID: 36321805 PMCID: PMC9635473 DOI: 10.1080/10717544.2022.2139431] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Acute liver injury is a common clinical disease, which easily leads to liver failure and endangers life, seriously threatening human health. Naringenin is a natural flavonoid that holds therapeutic potential against various liver injuries; however it has poor water solubility and bioavailability. In this study, we aimed to develop naringenin-loaded bovine serum albumin nanoparticles (NGNPs) and to evaluate their hepatoprotective effect and underlying mechanisms against acetaminophen overdose toxicity. In vitro data indicated that NGNPs significantly increased the drug solubility and also more effectively protected the hepatocyte cells from oxidative damage during hydrogen peroxide exposure or lipopolysaccharide (LPS) stimulation. In vivo results confirmed that NGNPs showed an enhanced accumulation in the liver tissue. In the murine model of acetaminophen-induced hepatotoxicity, NGNPs could effectively alleviate the progression of acute liver injury by reducing drug overdose-induced levels of oxidative stress, inflammation and apoptosis in hepatocytes. In conclusion, NGNPs has strong hepatoprotective effects against acetaminophen induced acute liver injury.
Collapse
Affiliation(s)
- Ruijie Lu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China,The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Run-Jie Yu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chunhui Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qian Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yunxia Xuan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zeqing Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhimin He
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yan Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Longfa Kou
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Ying-Zheng Zhao
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Qing Yao
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China,CONTACT Qing Yao Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, University Town, Chashan, Wenzhou 325000, Zhejiang, China
| | - Shi-Hao Xu
- Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Shi-Hao Xu Department of Ultrasonography, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
15
|
Wang Y, Iqbal H, Ur-Rehman U, Zhai L, Yuan Z, Razzaq A, Lv M, Wei H, Ning X, Xin J, Xiao R. Albumin-based nanodevices for breast cancer diagnosis and therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Huang D, Gui J, Chen X, Yu R, Gong T, Zhang Z, Fu Y. Chondroitin Sulfate-Derived Paclitaxel Nanocrystal via π-π Stacking with Enhanced Stability and Tumor Targetability. ACS APPLIED MATERIALS & INTERFACES 2022; 14:51776-51789. [PMID: 36350778 DOI: 10.1021/acsami.2c15881] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Nanocrystals with high drug loading have become a viable strategy for solubilizing drugs with poor aqueous solubility. It remains challenging, however, to synthesize nanocrystals with sufficient stability and targeting potential. Here, we report a novel nanocrystal platform synthesized using paclitaxel (PTX) and Fmoc-8-amino-3,6-dioxaoctanoic acid (Fmoc-AEEA)-conjugated chondroitin sulfate (CS) (CS-Fmoc) via π-π stacking to afford a stable formulation with CD44 targetability (PTX NC@CS-Fmoc). The PTX NC@CS-Fmoc exhibited rodlike shapes with an average hydrodynamic size of 173.6 ± 0.7 nm (PDI = 0.11 ± 0.04) and a drug loading of up to 31.3 ± 0.6%. Next, PTX NC@CS-Fmoc was subjected to lyophilization in the absence of cryoprotectants for long-term storage, and after redispersion, PTX NC@CS-Fmoc displayed an average hydrodynamic size of 205.3 ± 2.9 nm (PDI = 0.15 ± 0.01). In murine Panc02 cells, PTX NC@CS-Fmoc showed higher internalization efficiency than that of PTX nanocrystals without CS modification (PTX NC@F127) (P < 0.05) or that of CS-Fmoc micelles (P < 0.05). Moreover, PTX NC@CS-Fmoc appeared to accumulate in both lysosomes and Golgi apparatus, while CS-Fmoc micelles accumulated specifically in the Golgi apparatus. In the orthotopic Panc02 tumor-bearing mice model, PTX NC@CS-Fmoc showed higher tumor-specific accumulation than CS-Fmoc micelles, which also demonstrated comparable tumor growth inhibition as to Nab-PTX. Overall, the CS-Fmoc-derived nanocrystals represent a neat and viable formulation strategy for targeted chemotherapy with great potential for translational studies.
Collapse
Affiliation(s)
- Dandan Huang
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Jiajia Gui
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Xue Chen
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Ruilian Yu
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu610072, China
| | - Tao Gong
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Zhirong Zhang
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| | - Yao Fu
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu610041, China
| |
Collapse
|
17
|
Sukocheva OA, Liu J, Neganova ME, Beeraka NM, Aleksandrova YR, Manogaran P, Grigorevskikh EM, Chubarev VN, Fan R. Perspectives of using microRNA-loaded nanocarriers for epigenetic reprogramming of drug resistant colorectal cancers. Semin Cancer Biol 2022; 86:358-375. [PMID: 35623562 DOI: 10.1016/j.semcancer.2022.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/07/2023]
Abstract
Epigenetic regulation by microRNAs (miRs) demonstrated a promising therapeutic potential of these molecules to regulate genetic activity in different cancers, including colorectal cancers (CRCs). The RNA-based therapy does not change genetic codes in tumor cells but can silence oncogenes and/or reactivate inhibited tumor suppressor genes. In many cancers, specific miRs were shown to promote or stop tumor progression. Among confirmed and powerful epigenetic regulators of colon carcinogenesis and development of resistance are onco-miRs, which include let-7, miR-21, miR-22, miR-23a, miR-27a, miR-34, miR-92, miR-96, miR-125b, miR-135b, miR-182, miR-200c, miR-203, miR-221, miR-421, miR-451, and others. Moreover, various tumor-suppressor miRs (miR-15b-5b, miR-18a, miR-20b, miR-22, miR-96, miR-139-5p, miR-145, miR-149, miR-197, miR-199b, miR-203, miR-214, miR-218, miR-320, miR-375-3p, miR-409-3p, miR-450b-5p, miR-494, miR-577, miR-874, and others) were found silenced in drug-resistant CRCs. Re-expression of tumor suppressor miR is complicated by the chemical nature of miRs that are not long-lasting compounds and require protection from the enzymatic degradation. Several recent studies explored application of miRs using nanocarrier complexes. This study critically describes the most successfully tested nanoparticle complexes used for intracellular delivery of nuclear acids and miRs, including micelles, liposomes, inorganic and polymeric NPs, dendrimers, and aptamers. Nanocarriers shield incorporated miRs and improve the agent stability in circulation. Attachment of antibodies and/or specific peptide or ligands facilitates cell-targeted miR delivery. Addressing in vivo challenges, a broad spectrum of non-toxic materials has been tested and indicated reliable advantages of lipid-based (lipoplexes) and polymer-based liposomes. Recent cutting-edge developments indicated that lipid-based complexes with multiple cargo, including several miRs, are the most effective approach to eradicate drug-resistant tumors. Focusing on CRC-specific miRs, this review provides a guidance and insights towards the most promising direction to achieve dramatic reduction in tumor growth and metastasis using miR-nanocarrier complexes.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China; The National Centre for Neuroimmunology and Emerging Diseases, Menzies Health Institute, Griffith University, Queensland, Australia; Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA 5042, Australia.
| | - Junqi Liu
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka 142432, Russia
| | - Narasimha M Beeraka
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China; Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow 119991, Russia; Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical College, Mysuru, Karnataka, India
| | - Yulia R Aleksandrova
- Institute of Physiologically Active Compounds of the Russian Academy of Sciences, 1, Severnii pr., Chernogolovka 142432, Russia
| | - Prasath Manogaran
- Translational Research Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Ekaterina M Grigorevskikh
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow 119991, Russia
| | - Vladimir N Chubarev
- Department of Human Anatomy, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Street, Moscow 119991, Russia
| | - Ruitai Fan
- Cancer Center and Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China.
| |
Collapse
|
18
|
Kou L, Huang H, Tang Y, Sun M, Li Y, Wu J, Zheng S, Zhao X, Chen D, Luo Z, Zhang X, Yao Q, Chen R. Opsonized nanoparticles target and regulate macrophage polarization for osteoarthritis therapy: A trapping strategy. J Control Release 2022; 347:237-255. [PMID: 35489544 DOI: 10.1016/j.jconrel.2022.04.037] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is a chronic disease caused by joint inflammation. Its occurrence and development depend on a continuous inflammation environment. The activated M1 macrophages play a critical role in the inflammatory response of OA. Regulating the pro-inflammatory M1 to anti-inflammatory M2 macrophages in the OA articular cavity could be a rational strategy for OA treatment. It has been acknowledged that activated macrophages could proactively capture opsonized nanoparticles in the bloodstream and then accumulate into the reticuloendothelial system (RES) organs. Based on this fact, a trapping strategy is proposed, which transforms a normal nanoparticle into an opsonized attractant to target and regulate macrophage polarization. In this study, the opsonized nanoparticle (IgG/Bb@BRPL) had several key features, including an immunoglobulin IgG (the opsonized layer), an anti-inflammatory agent berberine (Bb), and an oxidative stress-responsive bilirubin grafted polylysine biomaterial (BR-PLL) for drug loading (the inner nanocore). In vitro studies confirmed that IgG/Bb@BRPL prefer to be phagocytosed by M1 macrophage, not M0. And the internalized IgG/Bb@BRPL effectively promoted macrophage polarization toward the M2 phenotype and protected nearby chondrocytes. In vivo studies suggested that IgG/Bb@BRPL significantly enhanced therapeutic outcomes by suppressing inflammation and promoting cartilage repair while not prolonging the retention period compared to non-opsonized counterparts. This proof-of-concept study provided a novel opsonization trapping strategy for OA drug delivery and treatment.
Collapse
Affiliation(s)
- Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Wenzhou key Laboratory of basic science and translational research of radiation oncology, Zhejiang 325027, China
| | - Huirong Huang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yingying Tang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Meng Sun
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yingtao Li
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jianing Wu
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shimin Zheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xinyu Zhao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Daosen Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zucheng Luo
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
19
|
Preparation of Doxorubicin-Loaded Carboxymethyl-β-Cyclodextrin/Chitosan Nanoparticles with Antioxidant, Antitumor Activities and pH-Sensitive Release. Mar Drugs 2022; 20:md20050278. [PMID: 35621929 PMCID: PMC9146362 DOI: 10.3390/md20050278] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 01/16/2023] Open
Abstract
In this study, chitosan nanoparticles (HF-CD NPs) were synthesized by an ionic gelation method using negatively charged carboxymethyl-β-cyclodextrin and positively charged 2-hydroxypropyltrimethyl ammonium chloride chitosan bearing folic acid. The surface morphology of HF-CD NPs was spherical or oval, and they possessed relatively small particle size (192 ± 8 nm) and positive zeta potential (+20 ± 2 mV). Meanwhile, doxorubicin (Dox) was selected as model drug to investigate the prepared nanoparticles’ potential to serve as a drug delivery carrier. The drug loading efficiency of drug-loaded nanoparticles (HF-Dox-CD NPs) was 31.25%. In vitro release profiles showed that Dox release of nanoparticles represented a pH-sensitive sustained and controlled release characteristic. At the same time, the antioxidant activity of nanoparticles was measured, and chitosan nanoparticles possessed good antioxidant activity and could inhibit the lipid peroxidation inside the cell and avoid material infection. Notably, CCK-8 assay testified that the nanoparticles were safe drug carriers and significantly enhanced the antitumor activity of Dox. The nanoparticles possessed good antioxidant activity, pH-sensitive sustained controlled release, enhanced antitumor activity, and could be expected to serve as a drug carrier in future with broad application prospects.
Collapse
|
20
|
Feng J, Xiang L, Fang C, Tan Y, Li Y, Gong T, Wu Q, Gong T, Zhang Z. Dual-Targeting of Tumor Cells and Tumor-Associated Macrophages by Palmitic Acid Modified Albumin Nanoparticles for Antitumor and Antimetastasis Therapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:14887-14902. [PMID: 35344323 DOI: 10.1021/acsami.1c23274] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor-associated macrophages (TAMs), the most abundant immune cells in the tumor microenvironment (TME), profoundly affect the occurrence and development of tumors. To overcome the common limitations of TAMs-targeted delivery systems, such as off-target toxicity, high cost, and transformation probability, we fabricated pirarubicin (THP)-loaded palmitic acid modified human serum albumin nanoparticles (THP-PSA NPs) for dual-targeting of tumor cells and TAMs via acidic secretory proteins rich in cysteine (SPARC) and scavenger receptor-A (SR-A), respectively. In vitro, the THP-PSA NPs exhibit stronger cytotoxicity against 4T1 and M2 macrophages compared with THP-loaded human serum albumin nanoparticles (THP-HSA NPs). In vivo, the infiltration of myeloid-derived suppressor cells (MDSCs) and the secretion of immunosuppressive cytokines significantly decrease after effective elimination of the TAMs through the THP-PSA NPs treatment; this is accompanied by an increase in the immunostimulatory cytokine expression level. Moreover, the antitumor and antimetastasis experimental results indicate that the tumor volumes in mice treated with the THP-PSA NPs are effectively controlled, resulting in an inhibition rate of 81.0% and almost no metastases in the lung tissues. Finally, in terms of biological safety, the THP-PSA NPs perform similar to THP-HSA NPs, causing no damage to the liver or kidney.
Collapse
Affiliation(s)
- Jiaxing Feng
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ling Xiang
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Changlong Fang
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yulu Tan
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yan Li
- Sichuan Institute for Drug Control NMPA Key Laboratory for Quality Control and Evaluation of Vaccines and Biological Products, Chengdu 611731, China
| | - Ting Gong
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Qingsi Wu
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Tao Gong
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- 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 Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Cheng Y, Sun R, He M, Zhang M, Hou X, Sun Y, Wang J, Xu J, He H, Wang H, Lan M, Zhao Y, Yang Y, Chen X, Gao F. Light-switchable diphtherin transgene system combined with losartan for triple negtative breast cancer therapy based on nano drug delivery system. Int J Pharm 2022; 618:121613. [PMID: 35217071 DOI: 10.1016/j.ijpharm.2022.121613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 02/10/2022] [Accepted: 02/20/2022] [Indexed: 10/19/2022]
Abstract
Breast cancer is a common malignancy in women. The abnormally dense collagen network in breast cancer forms a therapeutic barrier that hinders the penetration and anti-tumor effect of drugs. To overcome this hurdle, we adopted a therapeutic strategy to treat breast cancer which combined a light-switchable transgene system and losartan. The light-switchable transgene system could regulate expression of the diphtheria toxin A fragment (DTA) gene with a high on/off ratio under blue light and had great potential for spatiotemporally controllable gene expression. We developed a nanoparticle drug delivery system to achieve tumor microenvironment-responsive and targeted delivery of DTA-encoded plasmids (pDTA) to tumor sites via dual targeting to cluster of differentiation-44 and αvβ3 receptors. In vivo studies indicated that the combination of pDTA and losartan reduce the concentration of collagen type I from 5.9 to 1.9 µg/g and decreased the level of active transforming growth factor-β by 75.0% in tumor tissues. Moreover, deeper tumor penetration was achieved, tumor growth was inhibited, and the survival rate was increased. Our combination strategy provides a novel and practical method for clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Yi Cheng
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai, 200237, China; Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Rui Sun
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Muye He
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Miao Zhang
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xinyu Hou
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yuji Sun
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jie Wang
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiajun Xu
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Hai He
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Hongtao Wang
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuzheng Zhao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Yang
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China
| | - Xianjun Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, China.
| | - Feng Gao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai, 200237, China; Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
22
|
Lecot N, Dávila B, Sánchez C, Fernández M, González M, Cabral P, Cerecetto H, Glisoni R. Development and Evaluation of 2-Amino-7-Fluorophenazine 5,10-Dioxide Polymeric Micelles as Antitumoral Agents for 4T1 Breast Cancer. Polymers (Basel) 2021; 14:71. [PMID: 35012094 PMCID: PMC8747360 DOI: 10.3390/polym14010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 11/23/2022] Open
Abstract
2-Amino-7-fluorophenazine 5,10-dioxide (FNZ) is a bioreducible prodrug, poorly soluble in water, with potential anticancer activity on hypoxic-tumors. This poor solubility limits its potential applications in clinic. Amphiphilic pristine polymeric micelles (PMs) based on triblock copolymers Pluronic® and Tetronic®, glycosylated derivatives and their mixtures with preformed-liposomes (LPS), were analyzed as strategies to improve the bioavailability of FNZ. FNZ encapsulations were performed and the obtaining nanostructures were characterized using UV-visible spectroscopy (UV-VIS), Transmission Electron Microscopy (TEM) and Dynamic Light Scattering (DLS). The most promising nanoformulations were analyzed for their potential toxicity and pharmacologically, at 20 mg/kg FNZ-doses, in a stage-IV murine metastatic-breast tumor model. The results revealed that the solubility of the encapsulated-FNZ increased up to 14 times and the analysis (UV-VIS, DLS and TEM) confirmed the interaction between vehicles and FNZ. In all the cases appropriate encapsulation efficiencies (greater than 75%), monodisperse nanometric particle sizes (PDI = 0.180-0.335), adequate Z-potentials (-1.59 to -26.4 mV), stabilities and spherical morphologies were obtained. The in vitro profile of FNZ controlled releases corresponded mainly to a kinetic Higuchi model. The in vitro/in vivo biological studies revealed non-toxicity and relevant tumor-weight diminution (up to 61%).
Collapse
Affiliation(s)
- Nicole Lecot
- Laboratorio de Radiofarmacia, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (P.C.); (H.C.)
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (B.D.); (C.S.); (M.G.)
| | - Belén Dávila
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (B.D.); (C.S.); (M.G.)
| | - Carina Sánchez
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (B.D.); (C.S.); (M.G.)
| | - Marcelo Fernández
- Laboratorio de Experimentación Animal, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay;
| | - Mercedes González
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (B.D.); (C.S.); (M.G.)
| | - Pablo Cabral
- Laboratorio de Radiofarmacia, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (P.C.); (H.C.)
| | - Hugo Cerecetto
- Laboratorio de Radiofarmacia, Centro de Investigaciones Nucleares, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (P.C.); (H.C.)
- Grupo de Química Orgánica Medicinal, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, Mataojo 2055, Montevideo 11400, Uruguay; (B.D.); (C.S.); (M.G.)
| | - Romina Glisoni
- Departamento de Tecnología Farmacéutica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, Buenos Aires C1113AAD, Argentina
- Instituto de Nanobiotecnología (NANOBIOTEC), CONICET-Universidad de Buenos Aires, Junín 956, Buenos Aires C1113AAD, Argentina
| |
Collapse
|
23
|
Demirci S, Sahiner M, Ari B, Sunol AK, Sahiner N. Chondroitin Sulfate-Based Cryogels for Biomedical Applications. Gels 2021; 7:127. [PMID: 34462411 PMCID: PMC8406096 DOI: 10.3390/gels7030127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/22/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022] Open
Abstract
Cryogels attained from natural materials offer exceptional properties in applications such as tissue engineering. Moreover, Halloysite Nanotubes (HNT) at 1:0.5 weight ratio were embedded into CS cryogels to render additional biomedical properties. The hemolysis index of CS cryogel and CS:HNT cryogels was calculated as 0.77 ± 0.41 and 0.81 ± 0.24 and defined as non-hemolytic materials. However, the blood coagulation indices of CS cryogel and CS:HNT cryogels were determined as 76 ± 2% and 68 ± 3%, suggesting a mild blood clotting capability. The maximum% swelling capacity of CS cryogel was measured as 3587 ± 186%, 4014 ± 184%, and 3984 ± 113%, at pH 1.0, pH 7.4 and pH 9.0, respectively, which were reduced to 1961 ± 288%, 2816 ± 192, 2405 ± 73%, respectively, for CS:HNT cryogel. It was found that CS cryogels can hydrolytically be degraded 41 ± 1% (by wt) in 16-day incubation, whereas the CS:HNT cryogels degraded by 30 ± 1 wt %. There is no chelation for HNT and 67.5 ± 1% Cu(II) chelation for linear CS was measured. On the other hand, the CS cryogel and CS:HNT cryogel revealed Cu(II) chelating capabilities of 60.1 ± 12.5%, and 43.2 ± 17.5%, respectively, from 0.1 mg/mL Cu(II) ion stock solution. Additionally, at 0.5 mg/mL CS, CS:HNT, and HNT, the Fe(II) chelation capacity of 99.7 ± 0.6, 86.2 ± 4.7% and only 11.9 ± 4.5% were measured, respectively, while no Fe(II) was chelated by linear CS chelated Fe(II). As the adjustable and controllable swelling properties of cryogels are important parameters in biomedical applications, the swelling properties of CS cryogels, at different solution pHs, e.g., at the solution pHs of 1.0, 7.4 and 9.0, were measured as 3587 ± 186%, 4014 ± 184%, and 3984 ± 113%, respectively, and the maximum selling% values of CS:HNT cryogels were determined as 1961 ± 288%, 2816 ± 192, 2405 ± 73%, respectively, at the same conditions. Alpha glucosidase enzyme interactions were investigated and found that CS-based cryogels can stimulate this enzyme at any CS formulation.
Collapse
Affiliation(s)
- Sahin Demirci
- Department of Chemistry, Faculty of Sciences & Arts, and Nanoscience and Technology Research and Application Center (NANORAC), Canakkale Onsekiz Mart University Terzioglu Campus, Canakkale 17100, Turkey; (S.D.); (B.A.)
| | - Mehtap Sahiner
- Faculty of Canakkale School of Applied Science, Canakkale Onsekiz Mart University Terzioglu Campus, Canakkale 17100, Turkey;
| | - Betul Ari
- Department of Chemistry, Faculty of Sciences & Arts, and Nanoscience and Technology Research and Application Center (NANORAC), Canakkale Onsekiz Mart University Terzioglu Campus, Canakkale 17100, Turkey; (S.D.); (B.A.)
| | - Aydin K. Sunol
- Department of Chemical & Biomedical Engineering, and Materials Science and Engineering, University of South Florida, Tampa, FL 33620, USA;
| | - Nurettin Sahiner
- Department of Chemistry, Faculty of Sciences & Arts, and Nanoscience and Technology Research and Application Center (NANORAC), Canakkale Onsekiz Mart University Terzioglu Campus, Canakkale 17100, Turkey; (S.D.); (B.A.)
- Department of Chemical & Biomedical Engineering, and Materials Science and Engineering, University of South Florida, Tampa, FL 33620, USA;
- Department of Ophthalmology, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|