1
|
Zhou L, Zhang TJ, Zhang L, Deng QY, Xia ZY, Chen SL, Cheng DB, Qiao ZY, Wang H. Stimuli-responsive peptide-based nanodrug delivery systems for tumor therapy. Chem Commun (Camb) 2025; 61:7384-7407. [PMID: 40293360 DOI: 10.1039/d5cc00950b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Compared to free chemotherapeutic drugs, nano-sized drug delivery systems exhibit enhanced therapeutic effects and reduced in vivo toxicity. Peptide-based drug delivery systems have garnered significant attention due to the advantageous properties of peptides, including their excellent biocompatibility, diverse side-chain functionalities, and ability to form stable secondary structures. Incorporating stimuli-responsive amino acid residues or specific responsive moieties within their side chains endows these peptide-based drug delivery systems with unique stimuli-responsive characteristics. In this review, we summarize recent advancements and mechanisms in peptide-based nanodrug delivery systems that are capable of responding to one or multiple stimuli as well as conclude with a concise overview of the challenges that lie ahead in this field.
Collapse
Affiliation(s)
- Lei Zhou
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Ting-Jie Zhang
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Lu Zhang
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Qiu-Ying Deng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Zhi-Yu Xia
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
| | - Si-Lin Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, Wuhan 430070, China.
- Hubei Longzhong Laboratory, Wuhan University of Technology Xiangyang Demonstration Zone, Xiangyang 441000, Hubei, China
| | - Zeng-Ying Qiao
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.
| |
Collapse
|
2
|
Firouzjaei AA, Mohammadi-Yeganeh S. Advancements in Targeted Therapies for Colorectal Cancer: Innovative Drug Formulation and Delivery Strategies. Arch Pharm (Weinheim) 2025; 358:e202400969. [PMID: 40259467 DOI: 10.1002/ardp.202400969] [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: 12/23/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/23/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality globally, with increasing incidence presenting significant treatment challenges. Traditional nontargeted therapies often result in high toxicity and limited efficacy, underscoring the need for improved treatment modalities. This review highlights recent advancements in drug delivery systems to enhance therapeutic outcomes for CRC. We examine innovative strategies, including computer-assisted pharmaceutical formulation, sustained-release matrices, and prodrugs, as well as targeted delivery mechanisms such as exosomes, liposomes, hydrogels, antibody-drug conjugates, and stimuli-responsive systems. These methodologies offer improved drug biodistribution, enhanced targeting of cancer cells, and reduced off-target effects, promising better clinical outcomes. Additionally, we discuss the development of novel formulations designed to optimize the delivery of therapeutic agents in advanced CRC. Ongoing clinical trials investigating these innovative systems signify a shift toward more effective patient treatment options. While challenges remain in the clinical application of these targeted therapies, continued research offers promising avenues for improving patient outcomes in CRC. This study aims to inform future strategies for managing this aggressive disease, ultimately enhancing survival rates and quality of life for affected individuals.
Collapse
Affiliation(s)
- Ali Ahmadizad Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Mao X, Li X, Liu S, Dong H, Jia W, Xu W, Wu X, Zhang Y. Global research trends on nanotechnology and colorectal cancer: a two-decade analysis. Nanomedicine (Lond) 2025; 20:343-354. [PMID: 39829306 PMCID: PMC11812397 DOI: 10.1080/17435889.2025.2452833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) remains a serious threat to humans worldwide. In this study, we used bibliometric analysis of the scientific literature to assess the trends and prospects of nanotechnology applications in CRC. MATERIALS AND METHODS We used the Web of Science Core Collection database to screen relevant publications on nanotechnology and CRC from 2004 to 2023 based on the inclusion criteria. Bibliometric analyses were performed on all selected publication and citation data. Visual analysis using VOSviewer and CiteSpace intuitively reflected the hotspots in this field. RESULTS In total, 2040 publications on nanotechnology in CRC were identified for this two-decade analysis. China (443, 29.14%) and the USA (199, 13.09%) were the top two most productive countries, and Sichuan University was the most prolific institution. The most influential journal was the International Journal of Nanomedicine. The author with the most papers was Li L. "Nanoparticles," "drug delivery," and "CRC" were the most common keywords. Green synthesis and gold nanoparticles were peripheral, incompletely developed topics. CONCLUSION This study provides a comprehensive overview of nanomaterials in CRC as clinical medicine, enriching the body of evidence in this field.
Collapse
Affiliation(s)
- Xinyu Mao
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinhao Li
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiwei Liu
- Medical School, Southeast University, Nanjing, China
| | - Hui Dong
- Medical School, Southeast University, Nanjing, China
| | - Weilu Jia
- Medical School, Southeast University, Nanjing, China
| | - Wenjing Xu
- Medical School, Southeast University, Nanjing, China
| | - Xia Wu
- Department of Microbiota Medicine, Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yewei Zhang
- Hepatopancreatobiliary Center, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Zeid LS, El-Masry HA, Mohamed HH, Hathout A, Younes AS, El-Kholy AA, Hamid AAMA, Elaziz NAA, Hafez FS, Mostafa MEE, Omar IMM, Ahmed TE, Darwish MSA. Review on macromolecule-based magnetic theranostic agents for biomedical applications: Targeted therapy and diagnostic imaging. J Pharm Sci 2025; 114:787-804. [PMID: 39710317 DOI: 10.1016/j.xphs.2024.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/24/2024]
Abstract
Clinical diagnostics and biological research are advanced by magnetic theranostic, which uses macromolecule-based magnetic theranostic agents for targeted therapy and diagnostic imaging. Within this review, the interaction of magnetic nanoparticles (MNPs) with biological macromolecules will be covered. The exciting potential of macromolecule-based magnetic theranostic agents to be used as a tool in drug delivery, photothermally therapy (PTT), gene therapy, hyperthermia therapy and photodynamic therapy (PDT) will be discussed. Innovative imaging technique: magnetic resonance imaging (MRI), magnetic particle imaging (MPI), fluorescence scanning, and photoacoustic scanning are revolutionizing biological diagnosis by potentially overcoming historical limitations. This review will cover the challenge of fabricating of macromolecule-based magnetic theranostic agents as a promising platform for theranostic that can combine therapies with diagnostics at subcellular level. Additionally, it looks at several chemical pathways leading to the process for generating MNPs, including the co-precipitation, the sol-gel, the hydrothermal synthesis, the polyol route, and the microemulsion technique. Eventually, the demands and prospects for magnetic theranostic are discussed, focusing on the requirement of further investigation to improve MNP structure towards biocompatible material and translation of these promising theranostic agents into clinical applications.
Collapse
Affiliation(s)
- Laila S Zeid
- Chemistry Department, Faculty of Science, Cairo University, Giza 12555, Egypt.
| | - Heba A El-Masry
- Chemistry & Microbiology Department, Faculty of Science, Menoufia University, Menoufia 32611, Egypt.
| | - Hend H Mohamed
- School of Biotechnology and Badr University in Cairo Research Center, Badr University in Cairo (BUC), Badr, Cairo 11829, Egypt
| | - Amira Hathout
- School of Biotechnology and Badr University in Cairo Research Center, Badr University in Cairo (BUC), Badr, Cairo 11829, Egypt.
| | - Ahmed S Younes
- Nuclear and Radiation Engineering Department, Faculty of Engineering, Alexandria University, Luxor 85951, Egypt.
| | - Aya A El-Kholy
- Applied Medical Chemistry Department, Medical Research Institute - Alexandria University, Alexandria 21561,Egypt.
| | - Aya Ashraf Muhammad Abdel Hamid
- Biotechnology Department Faculty of Agriculture, Ain shams university, 5 neighboring 33 investors May 15, Cairo 11724, Egypt.
| | - Naira Ali Abd Elaziz
- Department of Chemistry & Zoology, Faculty of Science, Cairo University, Mansouriya_Mansheat, Al Qanater Giza 12962 Egypt.
| | - Fatma Shaban Hafez
- Chemistry and Zoology Department, Faculty of Science, menofia university. Mahallet sobk, Ashmoun, Menofia 32839, Egypt.
| | - May Emad Eldin Mostafa
- Biotechnology Department, Faculty of Agriculture, Cairo University, Helmytelzyton, Cairo 11724, Egypt,.
| | - Islam M M Omar
- Biochemistry Department, Faculty of Veterinary Medicine, Zagazig University, El-agezy, Tanta 11113, Egypt.
| | - Tasneem Elsayed Ahmed
- Biochemistry Department, Faculty of Science, Ain shams university, Cairo 11595, Egypt.
| | - Mohamed S A Darwish
- Egyptian Petroleum Research Institute, 1 Ahmed El-Zomor Street, El Zohour Region, Nasr City, Cairo 11727, Egypt.
| |
Collapse
|
5
|
Xie L, Zuo X, Wang B, Li D, Chang W, Ji S, Ding D. Micelle-like Nanoparticles for Drug Delivery and Magnetically Enhanced Tumor Chemotherapy. ACS Biomater Sci Eng 2024; 10:7527-7538. [PMID: 39576943 DOI: 10.1021/acsbiomaterials.4c01897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2024]
Abstract
Using the coordination bonds between transition metal atoms and electron-rich functional groups, we synthesized two kinds of micelle-like nanoparticles. Using magnetic Fe3O4 as the core, poly(methyl methacrylate) (PMMA) and poly(acrylic acid) (PAA) brushes were grafted via activators regenerated by electron transfer for atom transfer radical polymerization (ARGET-ATRP), which formed micelle-like magnetic nanoparticles Fe3O4/PAA-PMMA with a hydrophobic outer layer and Fe3O4/PMMA-PAA with a hydrophilic outer layer. Both the micelle-like nanoparticles had amphiphilic properties and can be used to load hydrophilic or hydrophobic drugs. Even loaded with hydrophobic drugs, the micelle-like nanoparticles can still be dispersed in aqueous solution, and Fe3O4/PAA-PMMA had a higher loading content. As the drug carrier, these two micelle-like nanoparticles can be used for magnetically targeted drug delivery and magnetic resonance imaging due to superparamagnetic Fe3O4. In addition, due to the magnetic retention effect, the drug-loaded micelle-like nanoparticles remained at the tumor site, increasing the local drug concentration. At the same time, the drug-loaded micelle-like nanoparticles generated a magnetocaloric effect under the alternating magnetic field, which not only killed tumor cells by magnetic hyperthermia but also promoted the rapid release of drugs at the tumor site. In general, magnetically enhanced chemotherapy showed the best therapeutic effect on tumors.
Collapse
Affiliation(s)
- Liqin Xie
- School of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang 453003, China
| | - Xirui Zuo
- School of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang 453003, China
| | - Beilei Wang
- School of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang 453003, China
| | - Dan Li
- School of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang 453003, China
| | - Wenke Chang
- School of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang 453003, China
| | - Shenglu Ji
- School of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang 453003, China
| | - Dan Ding
- School of Life Science and Technology, Xinxiang Medical University, 601 Jinsui Road, Xinxiang 453003, China
- College of Life Sciences, Nankai University, 92 Weijin Road, Tianjin 300071, China
| |
Collapse
|
6
|
Perelló-Trias MT, Serrano-Muñoz AJ, Rodríguez-Fernández A, Segura-Sampedro JJ, Ramis JM, Monjo M. Intraperitoneal drug delivery systems for peritoneal carcinomatosis: Bridging the gap between research and clinical implementation. J Control Release 2024; 373:70-92. [PMID: 38986910 DOI: 10.1016/j.jconrel.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Several abdominal-located cancers develop metastasis within the peritoneum, what is called peritoneal carcinomatosis (PC), constituting a clinical challenge in their therapeutical management, often leading to poor prognoses. Current multidisciplinary strategies, including cytoreductive surgery (CRS), hyperthermic intraperitoneal chemotherapy (HIPEC), and pressurized intraperitoneal aerosol chemotherapy (PIPAC), demonstrate efficacy but have limitations. In response, alternative strategies are explored in the drug delivery field for intraperitoneal chemotherapy. Controlled drug delivery offers a promising avenue, maintaining localized drug concentrations for optimal PC management. Drug delivery systems (DDS), including hydrogels, implants, nanoparticles, and hybrid systems, show potential for sustained and region-specific drug release. The present review aims to offer an overview of the advances and current designs of DDS for PC chemotherapy administration, focusing on their composition, main characteristics, and principal experimental outcomes, highlighting the importance of biomaterial rationale design and in vitro/vivo models for their testing. Moreover, since clinical data for human subjects are scarce, we offer a critical discussion of the gap between bench and bedside in DDS translation, emphasizing the need for further research.
Collapse
Affiliation(s)
- M Teresa Perelló-Trias
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Antonio Jose Serrano-Muñoz
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Ana Rodríguez-Fernández
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain
| | - Juan José Segura-Sampedro
- Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; General & Digestive Surgery Service, Hospital Universitario La Paz, Paseo de la Castellana, 261, Fuencarral-El Pardo, 28046 Madrid, Spain; School of Medicine, University of the Balearic Islands (UIB), Carretera de Valldemossa, km 7,5, 07122 Palma, Balearic Islands, Spain
| | - Joana Maria Ramis
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain.
| | - Marta Monjo
- Cell Therapy and Tissue Engineering Group (TERCIT), Research Institute of Health Sciences (IUNICS), University of the Balearic Islands (UIB), Carretera de Valldemossa, Km 7,5, 07122 Palma, Balearic Islands, Spain; Health Research Institute of the Balearic Islands (IdISBa) - Carretera de Valldemossa, 79. Hospital Universitari Son Espases. Edifici S. 07120 Palma, Balearic Islands, Spain; Department of Fundamental Biology and Health Sciences, University of the Balearic Islands (UIB), Palma, Balearic Islands, Spain.
| |
Collapse
|
7
|
Song Y, Dong QQ, Ni YK, Xu XL, Chen CX, Chen W. Nano-Proteolysis Targeting Chimeras (Nano-PROTACs) in Cancer Therapy. Int J Nanomedicine 2024; 19:5739-5761. [PMID: 38882545 PMCID: PMC11180470 DOI: 10.2147/ijn.s448684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/30/2024] [Indexed: 06/18/2024] Open
Abstract
Proteolysis-targeting chimeras (PROTACs) are heterobifunctional molecules that have the capability to induce specific protein degradation. While playing a revolutionary role in effectively degrading the protein of interest (POI), PROTACs encounter certain limitations that impede their clinical translation. These limitations encompass off-target effects, inadequate cell membrane permeability, and the hook effect. The advent of nanotechnology presents a promising avenue to surmount the challenges associated with conventional PROTACs. The utilization of nano-proteolysis targeting chimeras (nano-PROTACs) holds the potential to enhance specific tissue accumulation, augment membrane permeability, and enable controlled release. Consequently, this approach has the capacity to significantly enhance the controllable degradation of target proteins. Additionally, they enable a synergistic effect by combining with other therapeutic strategies. This review comprehensively summarizes the structural basis, advantages, and limitations of PROTACs. Furthermore, it highlights the latest advancements in nanosystems engineered for delivering PROTACs, as well as the development of nano-sized PROTACs employing nanocarriers as linkers. Moreover, it delves into the underlying principles of nanotechnology tailored specifically for PROTACs, alongside the current prospects of clinical research. In conclusion, the integration of nanotechnology into PROTACs harbors vast potential in enhancing the anti-tumor treatment response and expediting clinical translation.
Collapse
Affiliation(s)
- Yue Song
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, People’s Republic of China
| | - Qing-Qing Dong
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Yi-Ke Ni
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Chao-Xiang Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang Province, 310015, People’s Republic of China
| | - Wei Chen
- ICU, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| |
Collapse
|
8
|
Szwed M, Marczak A. Application of Nanoparticles for Magnetic Hyperthermia for Cancer Treatment-The Current State of Knowledge. Cancers (Basel) 2024; 16:1156. [PMID: 38539491 PMCID: PMC10969623 DOI: 10.3390/cancers16061156] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 01/03/2025] Open
Abstract
Hyperthermia (HT) is an anti-cancer therapy commonly used with radio and chemotherapies based on applying heat (39-45 °C) to inhibit tumor growth. However, controlling heat towards tumors and not normal tissues is challenging. Therefore, nanoparticles (NPs) are used in HT to apply heat only to tumor tissues to induce DNA damage and the expression of heat shock proteins, which eventually result in apoptosis. The aim of this review article is to summarize recent advancements in HT with the use of magnetic NPs to locally increase temperature and promote cell death. In addition, the recent development of nanocarriers as NP-based drug delivery systems is discussed. Finally, the efficacy of HT combined with chemotherapy, radiotherapy, gene therapy, photothermal therapy, and immunotherapy is explored.
Collapse
Affiliation(s)
- Marzena Szwed
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143 St, 90-236 Lodz, Poland;
| | | |
Collapse
|
9
|
Wang T, Chang TMS. Superparamagnetic Artificial Cells PLGA-Fe 3O 4 Micro/Nanocapsules for Cancer Targeted Delivery. Cancers (Basel) 2023; 15:5807. [PMID: 38136352 PMCID: PMC10741498 DOI: 10.3390/cancers15245807] [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: 10/19/2023] [Revised: 11/10/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Artificial cells have been extensively used in many fields, such as nanomedicine, biotherapy, blood substitutes, drug delivery, enzyme/gene therapy, cancer therapy, and the COVID-19 vaccine. The unique properties of superparamagnetic Fe3O4 nanoparticles have contributed to increased interest in using superparamagnetic artificial cells (PLGA-Fe3O4 micro/nanocapsules) for targeted therapy. In this review, the preparation methods of Fe3O4 NPs and superparamagnetic artificial cell PLGA-drug-Fe3O4 micro/nanocapsules are discussed. This review also focuses on the recent progress of superparamagnetic PLGA-drug-Fe3O4 micro/nanocapsules as targeted therapeutics. We shall concentrate on the use of superparamagnetic artificial cells in the form of PLGA-drug-Fe3O4 nanocapsules for magnetic hyperthermia/photothermal therapy and cancer therapies, including lung breast cancer and glioblastoma.
Collapse
Affiliation(s)
| | - Thomas Ming Swi Chang
- Artificial Cells and Organs Research Centre, Departments of Medicine and Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
10
|
Fu Q, Shen S, Sun P, Gu Z, Bai Y, Wang X, Liu Z. Bioorthogonal chemistry for prodrug activation in vivo. Chem Soc Rev 2023; 52:7737-7772. [PMID: 37905601 DOI: 10.1039/d2cs00889k] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Prodrugs have emerged as a major strategy for addressing clinical challenges by improving drug pharmacokinetics, reducing toxicity, and enhancing treatment efficacy. The emergence of new bioorthogonal chemistry has greatly facilitated the development of prodrug strategies, enabling their activation through chemical and physical stimuli. This "on-demand" activation using bioorthogonal chemistry has revolutionized the research and development of prodrugs. Consequently, prodrug activation has garnered significant attention and emerged as an exciting field of translational research. This review summarizes the latest advancements in prodrug activation by utilizing bioorthogonal chemistry and mainly focuses on the activation of small-molecule prodrugs and antibody-drug conjugates. In addition, this review also discusses the opportunities and challenges of translating these advancements into clinical practice.
Collapse
Affiliation(s)
- Qunfeng Fu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
- Changping Laboratory, Beijing 102206, China
| | - Siyong Shen
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Pengwei Sun
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Zhi Gu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Yifei Bai
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Xianglin Wang
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
| | - Zhibo Liu
- Beijing National Laboratory for Molecular Sciences, Radiochemistry and Radiation Chemistry Key Laboratory of Fundamental Science, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China.
- Changping Laboratory, Beijing 102206, China
- Peking University-Tsinghua University Center for Life Sciences, Peking University, Beijing 100871, China
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
11
|
Chen X, Hou M, Zhang X, Liu H, Li W, Hong W. Active Targeted Janus Theranostic Nanoplatforms Enable Chemo-Photothermal Therapy to Inhibit the Growth of Breast Cancer. Mol Pharm 2023; 20:5800-5810. [PMID: 37822062 DOI: 10.1021/acs.molpharmaceut.3c00669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Nanoscale structures have been developed to serve various functions in cancer therapy, encompassing areas such as diagnosis, biomedical visualization, tissue regeneration, and drug delivery. Based on biocompatible chitosan oligosaccharides (COS) and gold nanorods (GNRs), we designed the drug delivery systems (GNR@polyacrylic acid-Mn@COS Janus nanoparticles (JNPs)), which achieved paclitaxel (PTX) loaded on the side of GNRs, and the PAA-Mn domain served as magnetic resonance imaging contrast agents. This system was found to be effectively delivered to tumor sites through the enhanced permeability and retention (EPR) effect and the active target of the COS. The uniform JNPs selectively targeted cancer cells instead of normal cells through interacting with the COS on the surface of tumor cells, and the pH/NIR-responsive drug release behavior further enhanced their therapeutic effects. The in vivo effects of JNPs against tumors were evaluated using subcutaneous and orthotopic lung metastasis models, yielding promising outcomes for both tumor diagnosis and cancer treatment. In conclusion, the obtained JNPs hold great promise as a theranostic nanoplatform with synergistic chemotherapeutic and photothermal effects.
Collapse
Affiliation(s)
- Xiangjun Chen
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Mingyi Hou
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Xinzhong Zhang
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Haixin Liu
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Wenting Li
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| | - Wei Hong
- School of Pharmacy, Shandong New Drug Loading & Release Technology and Preparation Engineering Laboratory, Binzhou Medical University, 346 Guanhai Road, Yantai 264003, P. R. China
| |
Collapse
|
12
|
Vijayan V, Sundaram A, Vasukutty A, Bardhan R, Uthaman S, Park IK. Tumor-targeting cell membrane-coated nanorings for magnetic-hyperthermia-induced tumor ablation. Biomater Sci 2023; 11:7188-7202. [PMID: 37750339 PMCID: PMC10595517 DOI: 10.1039/d3bm01141k] [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: 07/09/2023] [Accepted: 09/10/2023] [Indexed: 09/27/2023]
Abstract
Magnetic hyperthermia has attracted considerable attention for efficient cancer therapy because of its noninvasive nature, deep tissue penetration, and minimal damage to healthy tissues. Herein, we have fused cancer cell membrane fragments with lipids and cloaked them on magnetic nanorings to form targeted Fe nanorings (TF) for tumor-targeted magnetic hyperthermia-induced tumor ablation. In our approach, cell membrane fragments from cancer cells were fused with lipids to form vesicles, which could efficiently encapsulate magnetic nanorings, thereby forming TF. We observed that TF have high tumor uptake via homotypic targeting, where cancer cells take up TF through membrane fusion. Under an external alternating magnetic field (AMF), TF accumulated in the tumors are heated, driving magnetic-hyperthermia-induced tumor cell death. Our in vitro studies show that self-targeting TF efficiently localized in cancer cells and induced cell death with an AMF, which was shown by a live/dead assay. Our findings demonstrate the potential of TF in tumor ablation, thereby making them promising and efficient nanosystems for tumor-targeted theranostics.
Collapse
Affiliation(s)
- Veena Vijayan
- Department of Biomedical Sciences, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun, Jeonnam 58128, Republic of Korea.
| | - Aravindkumar Sundaram
- Department of Biomedical Sciences, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun, Jeonnam 58128, Republic of Korea.
| | - Arathy Vasukutty
- Department of Biomedical Sciences, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun, Jeonnam 58128, Republic of Korea.
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50012, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA.
| | - Saji Uthaman
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50012, USA
- Nanovaccine Institute, Iowa State University, Ames, IA 50012, USA.
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, 264 Seoyang-ro, Hwasun, Jeonnam 58128, Republic of Korea.
| |
Collapse
|
13
|
Gago L, Quiñonero F, Perazzoli G, Melguizo C, Prados J, Ortiz R, Cabeza L. Nanomedicine and Hyperthermia for the Treatment of Gastrointestinal Cancer: A Systematic Review. Pharmaceutics 2023; 15:1958. [PMID: 37514144 PMCID: PMC10386177 DOI: 10.3390/pharmaceutics15071958] [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: 06/09/2023] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The incidence of gastrointestinal cancers has increased in recent years. Current treatments present numerous challenges, including drug resistance, non-specificity, and severe side effects, needing the exploration of new therapeutic strategies. One promising avenue is the use of magnetic nanoparticles, which have gained considerable interest due to their ability to generate heat in tumor regions upon the application of an external alternating magnetic field, a process known as hyperthermia. This review conducted a systematic search of in vitro and in vivo studies published in the last decade that employ hyperthermia therapy mediated by magnetic nanoparticles for treating gastrointestinal cancers. After applying various inclusion and exclusion criteria (studies in the last 10 years where hyperthermia using alternative magnetic field is applied), a total of 40 articles were analyzed. The results revealed that iron oxide is the preferred material for magnetism generation in the nanoparticles, and colorectal cancer is the most studied gastrointestinal cancer. Interestingly, novel therapies employing nanoparticles loaded with chemotherapeutic drugs in combination with magnetic hyperthermia demonstrated an excellent antitumor effect. In conclusion, hyperthermia treatments mediated by magnetic nanoparticles appear to be an effective approach for the treatment of gastrointestinal cancers, offering advantages over traditional therapies.
Collapse
Affiliation(s)
- Lidia Gago
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Francisco Quiñonero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Raul Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| | - Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Biosanitary Institute of Granada (ibs.GRANADA), SAS-University of Granada, 18014 Granada, Spain
| |
Collapse
|
14
|
Yusefi M, Shameli K, Jahangirian H, Teow SY, Afsah-Hejri L, Mohamad Sukri SNA, Kuča K. How Magnetic Composites are Effective Anticancer Therapeutics? A Comprehensive Review of the Literature. Int J Nanomedicine 2023; 18:3535-3575. [PMID: 37409027 PMCID: PMC10319292 DOI: 10.2147/ijn.s375964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 05/31/2023] [Indexed: 07/07/2023] Open
Abstract
Chemotherapy is the most prominent route in cancer therapy for prolonging the lifespan of cancer patients. However, its non-target specificity and the resulting off-target cytotoxicities have been reported. Recent in vitro and in vivo studies using magnetic nanocomposites (MNCs) for magnetothermal chemotherapy may potentially improve the therapeutic outcome by increasing the target selectivity. In this review, magnetic hyperthermia therapy and magnetic targeting using drug-loaded MNCs are revisited, focusing on magnetism, the fabrication and structures of magnetic nanoparticles, surface modifications, biocompatible coating, shape, size, and other important physicochemical properties of MNCs, along with the parameters of the hyperthermia therapy and external magnetic field. Due to the limited drug-loading capacity and low biocompatibility, the use of magnetic nanoparticles (MNPs) as drug delivery system has lost traction. In contrast, MNCs show higher biocompatibility, multifunctional physicochemical properties, high drug encapsulation, and multi-stages of controlled release for localized synergistic chemo-thermotherapy. Further, combining various forms of magnetic cores and pH-sensitive coating agents can generate a more robust pH, magneto, and thermo-responsive drug delivery system. Thus, MNCs are ideal candidate as smart and remotely guided drug delivery system due to a) their magneto effects and guide-ability by the external magnetic fields, b) on-demand drug release performance, and c) thermo-chemosensitization under an applied alternating magnetic field where the tumor is selectively incinerated without harming surrounding non-tumor tissues. Given the important effects of synthesis methods, surface modifications, and coating of MNCs on their anticancer properties, we reviewed the most recent studies on magnetic hyperthermia, targeted drug delivery systems in cancer therapy, and magnetothermal chemotherapy to provide insights on the current development of MNC-based anticancer nanocarrier.
Collapse
Affiliation(s)
- Mostafa Yusefi
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Kuala Lumpur, Malaysia
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Kamyar Shameli
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, 81675, Germany
| | | | - Sin-Yeang Teow
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, 325060, People’s Republic of China
| | - Leili Afsah-Hejri
- Department of Food Safety and Quality, School of Business, Science and Technology, Lakeland University Plymouth, WI 53073, USA
| | | | - Kamil Kuča
- Malaysia-Japan International Institute of Technology, Universiti Teknologi Malaysia, Kuala Lumpur, Malaysia
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
15
|
Orel VB, Papazoglou ΑS, Tsagkaris C, Moysidis DV, Papadakos S, Galkin OY, Orel VE, Syvak LA. Nanotherapy based on magneto-mechanochemical modulation of tumor redox state. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1868. [PMID: 36289050 DOI: 10.1002/wnan.1868] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/25/2022] [Accepted: 10/10/2022] [Indexed: 05/13/2023]
Abstract
Magnetic nanoparticles (MNs) are typically used as contrast agents for magnetic resonance imaging or as drug carriers with a remotely controlled delivery to the tumor. However, they can also potentiate the action of anticancer drugs under the influence of applied constant magnetic (CMFs) and electromagnetic fields (EMFs). This review demonstrates the role of magneto-mechanochemical effects produced by MNs alone and loaded with anticancer agents (MNCs) in response to CMFs and EMFs for modulation of tumor redox state. The combined treatment is suggested to act by two mechanisms: spin-dependent electron transport propagates free radical chain reactions, while magnetomechanical interactions cause conformational changes in drug molecules loaded onto MNs and generate reactive oxygen species (ROS). By adjusting the parameters of CMFs and EMFs during the magneto-mechanochemical synthesis and subsequent treatment, it is possible to modulate ROS production and switch redox signaling involved in ERK1/2 and NF-κB pathways from initiation of tumor growth to inhibition. Observations of tumor volume in different animal models and treatment combinations reported a 6%-70% reduction as compared with conventional drugs. Despite these results, there is a general lack of research in magnetic nanotheranostics that link redox changes across multiple levels of organization in the tumor-bearing host. Further multidisciplinary studies with more focus on the relationship between the electron transport processes in biomolecules and their effects on the tumor-host interaction should accelerate the clinical translation of magnetic nanotheranostics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Valerii B Orel
- National Cancer Institute, Kyiv, Ukraine
- Faculty of Biomedical Engineering, National Technical University of Ukraine "Igor Sikorsky Kyiv Polytechnic Institute", Kyiv, Ukraine
| | | | - Christos Tsagkaris
- Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia
| | - Dimitrios V Moysidis
- Department of Cardiology, Hippokration General Hospital of Thessaloniki, Thessaloniki, Greece
| | | | - Olexander Yu Galkin
- Faculty of Biomedical Engineering, National Technical University of Ukraine "Igor Sikorsky Kyiv Polytechnic Institute", Kyiv, Ukraine
| | - Valerii E Orel
- National Cancer Institute, Kyiv, Ukraine
- Faculty of Biomedical Engineering, National Technical University of Ukraine "Igor Sikorsky Kyiv Polytechnic Institute", Kyiv, Ukraine
| | | |
Collapse
|
16
|
Bag N, Bardhan S, Roy S, Roy J, Mondal D, Guo B, Das S. Nanoparticle-mediated stimulus-responsive antibacterial therapy. Biomater Sci 2023; 11:1994-2019. [PMID: 36748318 DOI: 10.1039/d2bm01941h] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The limitations associated with conventional antibacterial therapies and the subsequent amplification of multidrug-resistant (MDR) microorganisms have increased, necessitating the urgent development of innovative antibacterial techniques. Accordingly, nanoparticle-mediated therapeutics have emerged as potential candidates for antibacterial treatment due to their suitable dimensions, penetration capacity, and high efficiency in targeted drug delivery. However, although nanoparticle-based drug delivery systems have been demonstrated to be effective, they are limited by their overuse and unwanted side effects. Thus, to overcome these drawbacks, stimulus-responsive antibiotic delivery has been extended as a promising strategy for site-specific restricted drug exemption. Nano-formulations that are triggered by various stimuli, such as intrinsic, extrinsic, and bacterial stimuli, have been developed. Thus, by harnessing the physicochemical properties of various nanoparticles, the selective release of therapeutic cargoes can be achieved through the application of a variety of local stimuli such as light, sound, irradiation, pH, and magnetic field. In this review, we also highlight the progress and perspectives of stimulus-responsive combination therapy, with special emphasis on the eradication of MDR strains and biofilms. Hence, this review addresses the advancement and challenges in the applications of stimulus-responsive nanoparticles together with the various future prospects of this technique.
Collapse
Affiliation(s)
- Neelanjana Bag
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| | - Souravi Bardhan
- Department of Physics, Jadavpur University, Kolkata-700032, India. .,Department of Environmental Science, Netaji Nagar College for Women, Kolkata-700092, India
| | - Shubham Roy
- Department of Physics, Jadavpur University, Kolkata-700032, India. .,Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Jhilik Roy
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| | - Dhananjoy Mondal
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| | - Bing Guo
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology and School of Science, Harbin Institute of Technology, Shenzhen-518055, China.
| | - Sukhen Das
- Department of Physics, Jadavpur University, Kolkata-700032, India.
| |
Collapse
|
17
|
Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm (Beijing) 2023; 4:e187. [PMID: 36654533 PMCID: PMC9834710 DOI: 10.1002/mco2.187] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2023] Open
Abstract
Cancer is a complex disease associated with a combination of abnormal physiological process and exhibiting dysfunctions in multiple systems. To provide effective treatment and diagnosis for cancer, current treatment strategies simultaneously focus on various tumor targets. Based on the rapid development of nanotechnology, nanocarriers have been shown to exhibit excellent potential for cancer therapy. Compared with nanoparticles with single functions, multifunctional nanoparticles are believed to be more aggressive and potent in the context of tumor targeting. However, the development of multifunctional nanoparticles is not simply an upgraded version of the original function, but involves a sophisticated system with a proper backbone, optimized modification sites, simple preparation method, and efficient function integration. Despite this, many well-designed multifunctional nanoparticles with promising therapeutic potential have emerged recently. Here, to give a detailed understanding and analyzation of the currently developed multifunctional nanoparticles, their platform structures with organic or inorganic backbones were systemically generalized. We emphasized on the functionalization and modification strategies, which provide additional functions to the nanoparticle. We also discussed the application combination strategies that were involved in the development of nanoformulations with functional crosstalk. This review thus provides an overview of the construction strategies and application advances of multifunctional nanoparticles.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Xingmei Duan
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuan ProvinceChina
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
18
|
Hughes KA, Misra B, Maghareh M, Bobbala S. Use of stimulatory responsive soft nanoparticles for intracellular drug delivery. NANO RESEARCH 2023; 16:6974-6990. [PMID: 36685637 PMCID: PMC9840428 DOI: 10.1007/s12274-022-5267-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 05/24/2023]
Abstract
Drug delivery has made tremendous advances in the last decade. Targeted therapies are increasingly common, with intracellular delivery highly impactful and sought after. Intracellular drug delivery systems have limitations due to imprecise and non-targeted release profiles. One way this can be addressed is through using stimuli-responsive soft nanoparticles, which contain materials with an organic backbone such as lipids and polymers. The choice of biomaterial is essential for soft nanoparticles to be responsive to internal or external stimuli. The nanoparticle must retain its integrity and payload in non-targeted physiological conditions while responding to particular intracellular environments where payload release is desired. Multiple internal and external factors could stimulate the intracellular release of drugs from nanoparticles. Internal stimuli include pH, oxidation, and enzymes, while external stimuli include ultrasound, light, electricity, and magnetic fields. Stimulatory responsive soft nanoparticulate systems specifically utilized to modulate intracellular delivery of drugs are explored in this review.
Collapse
Affiliation(s)
- Krystal A. Hughes
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Maryam Maghareh
- Department of Clinical Pharmacy, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV 26505 USA
| |
Collapse
|
19
|
Pradhan R, Dey A, Taliyan R, Puri A, Kharavtekar S, Dubey SK. Recent Advances in Targeted Nanocarriers for the Management of Triple Negative Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15010246. [PMID: 36678877 PMCID: PMC9866847 DOI: 10.3390/pharmaceutics15010246] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a life-threatening form of breast cancer which has been found to account for 15% of all the subtypes of breast cancer. Currently available treatments are significantly less effective in TNBC management because of several factors such as poor bioavailability, low specificity, multidrug resistance, poor cellular uptake, and unwanted side effects being the major ones. As a rapidly growing field, nano-therapeutics offers promising alternatives for breast cancer treatment. This platform provides a suitable pathway for crossing biological barriers and allowing sustained systemic circulation time and an improved pharmacokinetic profile of the drug. Apart from this, it also provides an optimized target-specific drug delivery system and improves drug accumulation in tumor cells. This review provides insights into the molecular mechanisms associated with the pathogenesis of TNBC, along with summarizing the conventional therapy and recent advances of different nano-carriers for the management of TNBC.
Collapse
Affiliation(s)
- Rajesh Pradhan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Anuradha Dey
- Medical Research, R&D Healthcare Division, Emami Ltd., Kolkata 700056, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
- Correspondence: (R.T.); (S.K.D.); Tel.: +91-6378-364-745 (R.T.); +91-8239-703-734 (S.K.D.)
| | - Anu Puri
- RNA Structure and Design Section, RNA Biology Laboratory (RBL), Center for Cancer Research, National Cancer Institute—Frederick, Frederick, MD 21702, USA
| | - Sanskruti Kharavtekar
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
- Medical Research, R&D Healthcare Division, Emami Ltd., Kolkata 700056, India
- Correspondence: (R.T.); (S.K.D.); Tel.: +91-6378-364-745 (R.T.); +91-8239-703-734 (S.K.D.)
| |
Collapse
|
20
|
Mohapatra A, Park IK. Recent Advances in ROS-Scavenging Metallic Nanozymes for Anti-Inflammatory Diseases: A Review. Chonnam Med J 2023; 59:13-23. [PMID: 36794252 PMCID: PMC9900225 DOI: 10.4068/cmj.2023.59.1.13] [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: 12/03/2022] [Revised: 12/26/2022] [Accepted: 12/27/2022] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress and dysregulated inflammatory responses are the hallmarks of inflammatory disorders, which are key contributors to high mortality rates and impose a substantial economic burden on society. Reactive oxygen species (ROS) are vital signaling molecules that promote the development of inflammatory disorders. The existing mainstream therapeutic approaches, including steroid and non-steroidal anti-inflammatory drugs, and proinflammatory cytokine inhibitors with anti-leucocyte inhibitors, are not efficient at curing the adverse effects of severe inflammation. Moreover, they have serious side effects. Metallic nanozymes (MNZs) mimic the endogenous enzymatic process and are promising candidates for the treatment of ROS-associated inflammatory disorders. Owing to the existing level of development of these metallic nanozymes, they are efficient at scavenging excess ROS and can resolve the drawbacks of traditional therapies. This review summarizes the context of ROS during inflammation and provides an overview of recent advances in metallic nanozymes as therapeutic agents. Furthermore, the challenges associated with MNZs and an outline for future to promote the clinical translation of MNZs are discussed. Our review of this expanding multidisciplinary field will benefit the current research and clinical application of metallic-nanozyme-based ROS scavenging in inflammatory disease treatment.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea
| | - In-Kyu Park
- Department of Biomedical Science, BK21 PLUS Center for Creative Biomedical Scientists, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
21
|
Swetha KL, Maravajjala KS, Li SD, Singh MS, Roy A. Breaking the niche: multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv Transl Res 2023; 13:105-134. [PMID: 35697894 DOI: 10.1007/s13346-022-01194-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Most of the current antitumor therapeutics were developed targeting the cancer cells only. Unfortunately, in the majority of tumors, this single-dimensional therapy is found to be ineffective. Advanced research has shown that cancer is a multicellular disorder. The tumor microenvironment (TME), which is made by a complex network of the bulk tumor cells and other supporting cells, plays a crucial role in tumor progression. Understanding the importance of the TME in tumor growth, different treatment modalities have been developed targeting these supporting cells. Recent clinical results suggest that simultaneously targeting multiple components of the tumor ecosystem with drug combinations can be highly effective. This type of "multidimensional" therapy has a high potential for cancer treatment. However, tumor-specific delivery of such multi-drug combinations remains a challenge. Nanomedicine could be utilized for the tumor-targeted delivery of such multidimensional therapeutics. In this review, we first give a brief overview of the major components of TME. We then highlight the latest developments in nanoparticle-based combination therapies, where one drug targets cancer cells and other drug targets tumor-supporting components in the TME for a synergistic effect. We include the latest preclinical and clinical studies and discuss innovative nanoparticle-mediated targeting strategies.
Collapse
Affiliation(s)
- K Laxmi Swetha
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Kavya Sree Maravajjala
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Westbrook Mall, Vancouver, BC, Canada
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, 201310, India. .,Center of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Aniruddha Roy
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
22
|
Wang C, Xu P, Li X, Zheng Y, Song Z. Research progress of stimulus-responsive antibacterial materials for bone infection. Front Bioeng Biotechnol 2022; 10:1069932. [PMID: 36636700 PMCID: PMC9831006 DOI: 10.3389/fbioe.2022.1069932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Infection is one of the most serious complications harmful to human health, which brings a huge burden to human health. Bone infection is one of the most common and serious complications of fracture and orthopaedic surgery. Antibacterial treatment is the premise of bone defect healing. Among all the antibacterial strategies, irritant antibacterial materials have unique advantages and the ability of targeted therapy. In this review, we focus on the research progress of irritating materials, the development of antibacterial materials and their advantages and disadvantages potential applications in bone infection.
Collapse
Affiliation(s)
| | | | | | - Yuhao Zheng
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| | - Zhiming Song
- Department of Sports Medicine, Orthopaedic Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Yang G, Liu Y, Chen J, Ding J, Chen X. Self-Adaptive Nanomaterials for Rational Drug Delivery in Cancer Therapy. ACCOUNTS OF MATERIALS RESEARCH 2022; 3:1232-1247. [DOI: 10.1021/accountsmr.2c00147] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Affiliation(s)
- Guanqing Yang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
| | - Yang Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Jinjin Chen
- 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, 107 Yanjiang West Road, Guangzhou 510120, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei 230026, P. R. China
| |
Collapse
|
24
|
Armenia I, Cuestas Ayllón C, Torres Herrero B, Bussolari F, Alfranca G, Grazú V, Martínez de la Fuente J. Photonic and magnetic materials for on-demand local drug delivery. Adv Drug Deliv Rev 2022; 191:114584. [PMID: 36273514 DOI: 10.1016/j.addr.2022.114584] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 10/16/2022] [Indexed: 02/06/2023]
Abstract
Nanomedicine has been considered a promising tool for biomedical research and clinical practice in the 21st century because of the great impact nanomaterials could have on human health. The generation of new smart nanomaterials, which enable time- and space-controlled drug delivery, improve the limitations of conventional treatments, such as non-specific targeting, poor biodistribution and permeability. These smart nanomaterials can respond to internal biological stimuli (pH, enzyme expression and redox potential) and/or external stimuli (such as temperature, ultrasound, magnetic field and light) to further the precision of therapies. To this end, photonic and magnetic nanoparticles, such as gold, silver and iron oxide, have been used to increase sensitivity and responsiveness to external stimuli. In this review, we aim to report the main and most recent systems that involve photonic or magnetic nanomaterials for external stimulus-responsive drug release. The uniqueness of this review lies in highlighting the versatility of integrating these materials within different carriers. This leads to enhanced performance in terms of in vitro and in vivo efficacy, stability and toxicity. We also point out the current regulatory challenges for the translation of these systems from the bench to the bedside, as well as the yet unresolved matter regarding the standardization of these materials.
Collapse
Affiliation(s)
- Ilaria Armenia
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain.
| | - Carlos Cuestas Ayllón
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Beatriz Torres Herrero
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Francesca Bussolari
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Gabriel Alfranca
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain
| | - Valeria Grazú
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain; Centro de Investigación Biomédica em Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| | - Jesús Martínez de la Fuente
- BioNanoSurf Group, Instituto de Nanociencia y Materiales de Aragón (INMA,CSIC-UNIZAR), Edificio I +D, 50018 Zaragoza, Spain; Centro de Investigación Biomédica em Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
25
|
Wang Y, Liu L, Le Z, Tay A. Analysis of Nanomedicine Efficacy for Osteoarthritis. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Yuwen Wang
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Ling Liu
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
| | - Zhicheng Le
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Andy Tay
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
- Institute of Health Innovation and Technology National University of Singapore Singapore 117599 Singapore
- Tissue Engineering Programme National University of Singapore Singapore 117510 Singapore
| |
Collapse
|
26
|
Akhtar N, Mohammed HA, Yusuf M, Al-Subaiyel A, Sulaiman GM, Khan RA. SPIONs Conjugate Supported Anticancer Drug Doxorubicin's Delivery: Current Status, Challenges, and Prospects. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3686. [PMID: 36296877 PMCID: PMC9611558 DOI: 10.3390/nano12203686] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Considerable efforts have been directed towards development of nano-structured carriers to overcome the limitations of anticancer drug, doxorubicin's, delivery to various cancer sites. The drug's severe toxicity to cardio and hepatic systems, low therapeutic outcomes, inappropriate dose-demands, metastatic and general resistance, together with non-selectivity of the drug have led to the development of superparamagnetic iron oxide nanoparticles (SPIONs)-based drug delivery modules. Nano-scale polymeric co-encapsulation of the drug, doxorubicin, with SPIONs, the SPIONs surface end-groups' cappings with small molecular entities, as well as structural modifications of the SPIONs' surface-located functional end-groups, to attach the doxorubicin, have been achieved through chemical bonding by conjugation and cross-linking of natural and synthetic polymers, attachments of SPIONs made directly to the non-polymeric entities, and attachments made through mediation of molecular-spacer as well as non-spacer mediated attachments of several types of chemical entities, together with the physico-chemical bondings of the moieties, e.g., peptides, proteins, antibodies, antigens, aptamers, glycoproteins, and enzymes, etc. to the SPIONs which are capable of targeting multiple kinds of cancerous sites, have provided stable and functional SPIONs-based nano-carriers suitable for the systemic, and in vitro deliveries, together with being suitable for other biomedical/biotechnical applications. Together with the SPIONs inherent properties, and ability to respond to magnetic resonance, fluorescence-directed, dual-module, and molecular-level tumor imaging; as well as multi-modular cancer cell targeting; magnetic-field-inducible drug-elution capacity, and the SPIONs' magnetometry-led feasibility to reach cancer action sites have made sensing, imaging, and drug and other payloads deliveries to cancerous sites for cancer treatment a viable option. Innovations in the preparation of SPIONs-based delivery modules, as biocompatible carriers; development of delivery route modalities; approaches to enhancing their drug delivery-cum-bioavailability have explicitly established the SPIONs' versatility for oncological theranostics and imaging. The current review outlines the development of various SPIONs-based nano-carriers for targeted doxorubicin delivery to different cancer sites through multiple methods, modalities, and materials, wherein high-potential nano-structured platforms have been conceptualized, developed, and tested for, both, in vivo and in vitro conditions. The current state of the knowledge in this arena have provided definite dose-control, site-specificity, stability, transport feasibility, and effective onsite drug de-loading, however, with certain limitations, and these shortcomings have opened the field for further advancements by identifying the bottlenecks, suggestive and plausible remediation, as well as more clear directions for future development.
Collapse
Affiliation(s)
- Naseem Akhtar
- Department of Pharmaceutics, College of Dentistry & Pharmacy, Buraydah Private Colleges, P.O. Box 31717, Buraydah 51418, Qassim, Saudi Arabia
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Qassim, Saudi Arabia
| | - Mohammed Yusuf
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Mecca, Saudi Arabia
| | - Amal Al-Subaiyel
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Qassim, Saudi Arabia
| | - Ghassan M. Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq
| | - Riaz A. Khan
- Department of Medicinal Chemistry & Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Qassim, Saudi Arabia
| |
Collapse
|
27
|
Nazli A, He DL, Liao D, Khan MZI, Huang C, He Y. Strategies and progresses for enhancing targeted antibiotic delivery. Adv Drug Deliv Rev 2022; 189:114502. [PMID: 35998828 DOI: 10.1016/j.addr.2022.114502] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 08/10/2022] [Accepted: 08/16/2022] [Indexed: 01/24/2023]
Abstract
Antibiotic resistance is a global health issue and a potential risk for society. Antibiotics administered through conventional formulations are devoid of targeting effect and often spread to various undesired body sites, leading to sub-lethal concentrations at the site of action and thus resulting in emergence of resistance, as well as side effects. Moreover, we have a very slim antibiotic pipeline. Drug-delivery systems have been designed to control the rate, time, and site of drug release, and innovative approaches for antibiotic delivery provide a glint of hope for addressing these issues. This review elaborates different delivery strategies and approaches employed to overcome the limitations of conventional antibiotic therapy. These include antibiotic conjugates, prodrugs, and nanocarriers for local and targeted antibiotic release. In addition, a wide range of stimuli-responsive nanocarriers and biological carriers for targeted antibiotic delivery are discussed. The potential advantages and limitations of targeted antibiotic delivery strategies are described along with possible solutions to avoid these limitations. A number of antibiotics successfully delivered through these approaches with attained outcomes and potentials are reviewed.
Collapse
Affiliation(s)
- Adila Nazli
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China
| | - David L He
- College of Chemistry, University of California, Berkeley, CA 94720, United States
| | - Dandan Liao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China
| | | | - Chao Huang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China.
| | - Yun He
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing 401331, PR China.
| |
Collapse
|
28
|
Singh R, Prasad A, Kumar B, Kumari S, Sahu RK, Hedau ST. Potential of Dual Drug Delivery Systems: MOF as Hybrid Nanocarrier for Dual Drug Delivery in Cancer Treatment. ChemistrySelect 2022. [DOI: 10.1002/slct.202201288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Ragini Singh
- Division of Molecular Oncology ICMR-National Institute of Cancer Prevention and Research I-7, Sector 39 Noida 201301 Gautam Budha Nagar, U.P. India
| | - Amrita Prasad
- Department of Chemistry Magadh Mahila College Patna University Patna Bihar. India
| | - Binayak Kumar
- Division of Molecular Oncology ICMR-National Institute of Cancer Prevention and Research I-7, Sector 39 Noida 201301 Gautam Budha Nagar, U.P. India
| | - Soni Kumari
- Division of Molecular Oncology ICMR-National Institute of Cancer Prevention and Research I-7, Sector 39 Noida 201301 Gautam Budha Nagar, U.P. India
| | - Ram Krishna Sahu
- Division of Molecular Oncology ICMR-National Institute of Cancer Prevention and Research I-7, Sector 39 Noida 201301 Gautam Budha Nagar, U.P. India
| | - Suresh T. Hedau
- Division of Molecular Oncology ICMR-National Institute of Cancer Prevention and Research I-7, Sector 39 Noida 201301 Gautam Budha Nagar, U.P. India
| |
Collapse
|
29
|
Veletić M, Apu EH, Simić M, Bergsland J, Balasingham I, Contag CH, Ashammakhi N. Implants with Sensing Capabilities. Chem Rev 2022; 122:16329-16363. [PMID: 35981266 DOI: 10.1021/acs.chemrev.2c00005] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Because of the aging human population and increased numbers of surgical procedures being performed, there is a growing number of biomedical devices being implanted each year. Although the benefits of implants are significant, there are risks to having foreign materials in the body that may lead to complications that may remain undetectable until a time at which the damage done becomes irreversible. To address this challenge, advances in implantable sensors may enable early detection of even minor changes in the implants or the surrounding tissues and provide early cues for intervention. Therefore, integrating sensors with implants will enable real-time monitoring and lead to improvements in implant function. Sensor integration has been mostly applied to cardiovascular, neural, and orthopedic implants, and advances in combined implant-sensor devices have been significant, yet there are needs still to be addressed. Sensor-integrating implants are still in their infancy; however, some have already made it to the clinic. With an interdisciplinary approach, these sensor-integrating devices will become more efficient, providing clear paths to clinical translation in the future.
Collapse
Affiliation(s)
- Mladen Veletić
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ehsanul Hoque Apu
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Division of Hematology and Oncology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Mitar Simić
- Faculty of Electrical Engineering, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Jacob Bergsland
- The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Ilangko Balasingham
- Department of Electronic Systems, Norwegian University of Science and Technology, 7491 Trondheim, Norway.,The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, 0372 Oslo, Norway
| | - Christopher H Contag
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ) and Department of Biomedical Engineering (BME), Michigan State University, East Lansing, Michigan 48824, United States.,Department of Bioengineering, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
30
|
Xiang X, Shi D, Gao J. The Advances and Biomedical Applications of Imageable Nanomaterials. Front Bioeng Biotechnol 2022; 10:914105. [PMID: 35866027 PMCID: PMC9294271 DOI: 10.3389/fbioe.2022.914105] [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: 04/06/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Nanomedicine shows great potential in screening, diagnosing and treating diseases. However, given the limitations of current technology, detection of some smaller lesions and drugs’ dynamic monitoring still need to be improved. With the advancement of nanotechnology, researchers have produced various nanomaterials with imaging capabilities which have shown great potential in biomedical research. Here, we summarized the researches based on the characteristics of imageable nanomaterials, highlighted the advantages and biomedical applications of imageable nanomaterials in the diagnosis and treatment of diseases, and discussed current challenges and prospects.
Collapse
Affiliation(s)
- Xiaohong Xiang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Doudou Shi
- Department of Gastroenterology, The Affiliated Hospital of Yan’an University, Yan’an, China
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Jianbo Gao,
| |
Collapse
|
31
|
De R, Mahata MK, Kim K. Structure-Based Varieties of Polymeric Nanocarriers and Influences of Their Physicochemical Properties on Drug Delivery Profiles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105373. [PMID: 35112798 PMCID: PMC8981462 DOI: 10.1002/advs.202105373] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/09/2022] [Indexed: 05/04/2023]
Abstract
Carriers are equally important as drugs. They can substantially improve bioavailability of cargos and safeguard healthy cells from toxic effects of certain therapeutics. Recently, polymeric nanocarriers (PNCs) have achieved significant success in delivering drugs not only to cells but also to subcellular organelles. Variety of natural sources, availability of different synthetic routes, versatile molecular architectures, exploitable physicochemical properties, biocompatibility, and biodegradability have presented polymers as one of the most desired materials for nanocarrier design. Recent innovative concepts and advances in PNC-associated nanotechnology are providing unprecedented opportunities to engineer nanocarriers and their functions. The efficiency of therapeutic loading has got considerably increased. Structural design-based varieties of PNCs are widely employed for the delivery of small therapeutic molecules to genes, and proteins. PNCs have gained ever-increasing attention and certainly paves the way to develop advanced nanomedicines. This article presents a comprehensive investigation of structural design-based varieties of PNCs and the influences of their physicochemical properties on drug delivery profiles with perspectives highlighting the inevitability of incorporating both the multi-stimuli-responsive and multi-drug delivery properties in a single carrier to design intelligent PNCs as new and emerging research directions in this rapidly developing area.
Collapse
Affiliation(s)
- Ranjit De
- Laboratory of Molecular NeurophysiologyDepartment of Life SciencesPohang University of Science and Technology (POSTECH)77 Cheongam‐RoPohangGyeongbuk37673South Korea
- Division of Integrative Biosciences and Biotechnology (IBB)Pohang University of Science and Technology (POSTECH)77 Cheongam‐RoPohangGyeongbuk37673South Korea
| | - Manoj Kumar Mahata
- Drittes Physikalisches Institut ‐ BiophysikGeorg‐August‐Universität GöttingenFriedrich‐Hund‐Platz 1Göttingen37077Germany
| | - Kyong‐Tai Kim
- Laboratory of Molecular NeurophysiologyDepartment of Life SciencesPohang University of Science and Technology (POSTECH)77 Cheongam‐RoPohangGyeongbuk37673South Korea
- Division of Integrative Biosciences and Biotechnology (IBB)Pohang University of Science and Technology (POSTECH)77 Cheongam‐RoPohangGyeongbuk37673South Korea
| |
Collapse
|
32
|
Zhang J, Lin Y, Lin Z, Wei Q, Qian J, Ruan R, Jiang X, Hou L, Song J, Ding J, Yang H. Stimuli-Responsive Nanoparticles for Controlled Drug Delivery in Synergistic Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103444. [PMID: 34927373 PMCID: PMC8844476 DOI: 10.1002/advs.202103444] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/28/2021] [Indexed: 05/10/2023]
Abstract
Cancer immunotherapy has achieved promising clinical progress over the recent years for its potential to treat metastatic tumors and inhibit their recurrences effectively. However, low patient response rates and dose-limiting toxicity remain as major dilemmas for immunotherapy. Stimuli-responsive nanoparticles (srNPs) combined with immunotherapy offer the possibility to amplify anti-tumor immune responses, where the weak acidity, high concentration of glutathione, overexpressions of enzymes, and reactive oxygen species, and external stimuli in tumors act as triggers for controlled drug release. This review highlights the design of srNPs based on tumor microenvironment and/or external stimuli to combine with different anti-tumor drugs, especially the immunoregulatory agents, which eventually realize synergistic immunotherapy of malignant primary or metastatic tumors and acquire a long-term immune memory to prevent tumor recurrence. The authors hope that this review can provide theoretical guidance for the construction and clinical transformation of smart srNPs for controlled drug delivery in synergistic cancer immunotherapy.
Collapse
Affiliation(s)
- Jin Zhang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Yandai Lin
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Zhe Lin
- Ruisi (Fujian) Biomedical Engineering Research Center Co LtdFuzhou350100P. R. China
| | - Qi Wei
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Jiaqi Qian
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Renjie Ruan
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Xiancai Jiang
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Linxi Hou
- Qingyuan Innovation LaboratoryCollege of Chemical EngineeringFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jibin Song
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- State Key Laboratory of Molecular Engineering of PolymersFudan University220 Handan RoadShanghai200433P. R. China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and BiologyState Key Laboratory of Photocatalysis on Energy and EnvironmentCollege of ChemistryFuzhou University2 Xueyuan RoadFuzhou350108P. R. China
| |
Collapse
|
33
|
Ryu C, Lee H, Kim H, Hwang S, Hadadian Y, Mohanty A, Park IK, Cho B, Yoon J, Lee JY. Highly Optimized Iron Oxide Embedded Poly(Lactic Acid) Nanocomposites for Effective Magnetic Hyperthermia and Biosecurity. Int J Nanomedicine 2022; 17:31-44. [PMID: 35023918 PMCID: PMC8743620 DOI: 10.2147/ijn.s344257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/21/2021] [Indexed: 12/29/2022] Open
Abstract
Introduction Iron oxide magnetic nanoparticles (IONPs) have attracted considerable attention for various biomedical applications owing to their ease of synthesis, strong magnetic properties, and biocompatibility. In particular, IONPs can generate heat under an alternating magnetic field, the effects of which have been extensively studied for magnetic hyperthermia therapy. However, the development of IONPs with high heating efficiency, biocompatibility, and colloidal stability in physiological environments is still required for their safe and effective application in biomedical fields. Methods We synthesized magnetic IONP/polymer nanocomposites (MNCs) by embedding IONPs in a poly(L-lactic acid) (PLA) matrix via nanoemulsion. The IONP contents (Fe: 9–22 [w/w]%) in MNCs were varied to investigate their effects on the magnetic and hyperthermia performances based on their optimal interparticle interactions. Further, we explored the stability, cytocompatibility, biodistribution, and in vivo tissue compatibility of the MNCs. Results The MNCs showed enhanced heating efficiency with over two-fold increase compared to nonembedded bare IONPs. The relationship between the IONP content and heating performance in MNCs was nonmonotonous. The highest heating performance was obtained from MNC2, which contain 13% Fe (w/w), implying that interparticle interactions in MNCs can be optimized to achieve high heating performance. In addition, the MNCs exhibited good colloidal stability under physiological conditions and maintained their heating efficiency during 48 h of incubation in cell culture medium. Both in vitro and in vivo studies revealed excellent biocompatibility of the MNC. Conclusion Our nanocomposites, comprising biocompatible IONPs and PLA, display improved heating efficiency, good colloidal stability, and cytocompatibility, and thus will be beneficial for diverse biomedical applications, including magnetic hyperthermia for cancer treatment.
Collapse
Affiliation(s)
- Chiseon Ryu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hwangjae Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hohyeon Kim
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Seong Hwang
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Yaser Hadadian
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.,Research Center for Nanorobotics in Brain, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Ayeskanta Mohanty
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun-gun, Jeollanam-do, Republic of Korea
| | - Beongki Cho
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jungwon Yoon
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.,Research Center for Nanorobotics in Brain, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| |
Collapse
|
34
|
Jiao W, Zhang T, Peng M, Yi J, He Y, Fan H. Design of Magnetic Nanoplatforms for Cancer Theranostics. BIOSENSORS 2022; 12:38. [PMID: 35049666 PMCID: PMC8774163 DOI: 10.3390/bios12010038] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 05/04/2023]
Abstract
Cancer is the top cause of death globally. Developing smart nanomedicines that are capable of diagnosis and therapy (theranostics) in one-nanoparticle systems are highly desirable for improving cancer treatment outcomes. The magnetic nanoplatforms are the ideal system for cancer theranostics, because of their diverse physiochemical properties and biological effects. In particular, a biocompatible iron oxide nanoparticle based magnetic nanoplatform can exhibit multiple magnetic-responsive behaviors under an external magnetic field and realize the integration of diagnosis (magnetic resonance imaging, ultrasonic imaging, photoacoustic imaging, etc.) and therapy (magnetic hyperthermia, photothermal therapy, controlled drug delivery and release, etc.) in vivo. Furthermore, due to considerable variation among tumors and individual patients, it is a requirement to design iron oxide nanoplatforms by the coordination of diverse functionalities for efficient and individualized theranostics. In this article, we will present an up-to-date overview on iron oxide nanoplatforms, including both iron oxide nanomaterials and those that can respond to an externally applied magnetic field, with an emphasis on their applications in cancer theranostics.
Collapse
Affiliation(s)
- Wangbo Jiao
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an 710069, China; (W.J.); (T.Z.); (M.P.)
| | - Tingbin Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an 710069, China; (W.J.); (T.Z.); (M.P.)
| | - Mingli Peng
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an 710069, China; (W.J.); (T.Z.); (M.P.)
| | - Jiabao Yi
- Global Innovative Centre for Advanced Nanomaterials, School of Engineering, The University of Newcastle, Newcastle, NSW 2308, Australia;
| | - Yuan He
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an 710069, China; (W.J.); (T.Z.); (M.P.)
| | - Haiming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi’an 710069, China; (W.J.); (T.Z.); (M.P.)
| |
Collapse
|
35
|
Lafuente-Gómez N, Latorre A, Milán-Rois P, Rodriguez Diaz C, Somoza Á. Stimuli-responsive nanomaterials for cancer treatment: boundaries, opportunities and applications. Chem Commun (Camb) 2021; 57:13662-13677. [PMID: 34874370 DOI: 10.1039/d1cc05056g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Small molecule drugs, including most chemotherapies, are rapidly degraded and/or eliminated from the body, which is why high doses of these drugs are necessary, potentially producing toxic effects. Several types of nanoparticles loaded with anti-cancer drugs have been designed to overcome the disadvantages of conventional therapies. Modified nanoparticles can circulate for a long time, thus improving the solubility and biodistribution of drugs. Furthermore, they also allow the controlled release of the payload once its target tissue has been reached. These mechanisms can reduce the exposure of healthy tissues to chemotherapeutics, since the drugs are only released in the presence of specific tumour stimuli. Overall, these properties can improve the effectiveness of treatments while reducing undesirable side effects. In this article, we review the recent advances in stimuli-responsive albumin, gold and magnetic nanostructures for controlled anti-cancer drug delivery. These nanostructures were designed to release drugs in response to different internal and external stimuli of the cellular environment, including pH, redox, light and magnetic fields. We also describe various examples of applications of these nanomaterials. Overall, we shed light on the properties, potential clinical translation and limitations of stimuli-responsive nanoparticles for cancer treatment.
Collapse
Affiliation(s)
- Nuria Lafuente-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Ana Latorre
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Paula Milán-Rois
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Ciro Rodriguez Diaz
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain.
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain. .,Unidad Asociada al Centro Nacional de Biotecnología (CSIC), 28049 Madrid, Spain
| |
Collapse
|
36
|
Hong Q, Huo S, Tang H, Qu X, Yue B. Smart Nanomaterials for Treatment of Biofilm in Orthopedic Implants. Front Bioeng Biotechnol 2021; 9:694635. [PMID: 34589470 PMCID: PMC8473796 DOI: 10.3389/fbioe.2021.694635] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 07/23/2021] [Indexed: 01/02/2023] Open
Abstract
Biofilms refer to complex bacterial communities that are attached to the surface of animate or inanimate objects, which highly resist the antibiotics or the host immune defense mechanisms. Pathogenic biofilms in medicine are general, chronic, and even costly, especially on medical devices and orthopedic implants. Bacteria within biofilms are the cause of many persistent infections, which are almost impossible to eradicate. Though some progress has been made in comprehending the mechanisms of biofilm formation and persistence, novel alternative compounds or strategies and effective anti-biofilm antibiotics are still lacking. Smart materials of nano size which are able to respond to an external stimulus or internal environment have a great range of applications in clinic. Recently, smart nanomaterials with or without carriage of antibiotics, targeting specific bacteria and biofilm under some stimuli, have shown great potential for pathogenic biofilm and resident bacteria eradication. First, this review briefly summarizes and describes the significance of biofilms and the process of biofilm formation. Then, we focus on some of the latest research studies involving biofilm elimination, which probably could be applied in orthopedic implants. Finally, some outstanding challenges and limitations that need to be settled urgently in order to make smart nanomaterials effectively target and treat implant biofilms are also discussed. It is hoped that there will be more novel anti-biofilm strategies for biofilm infection in the prospective future.
Collapse
Affiliation(s)
| | | | | | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopaedics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
37
|
Martins PM, Lima AC, Ribeiro S, Lanceros-Mendez S, Martins P. Magnetic Nanoparticles for Biomedical Applications: From the Soul of the Earth to the Deep History of Ourselves. ACS APPLIED BIO MATERIALS 2021; 4:5839-5870. [PMID: 35006927 DOI: 10.1021/acsabm.1c00440] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Precisely engineered magnetic nanoparticles (MNPs) have been widely explored for applications including theragnostic platforms, drug delivery systems, biomaterial/device coatings, tissue engineering scaffolds, performance-enhanced therapeutic alternatives, and even in SARS-CoV-2 detection strips. Such popularity is due to their unique, challenging, and tailorable physicochemical/magnetic properties. Given the wide biomedical-related potential applications of MNPs, significant achievements have been reached and published (exponentially) in the last five years, both in synthesis and application tailoring. Within this review, and in addition to essential works in this field, we have focused on the latest representative reports regarding the biomedical use of MNPs including characteristics related to their oriented synthesis, tailored geometry, and designed multibiofunctionality. Further, actual trends, needs, and limitations of magnetic-based nanostructures for biomedical applications will also be discussed.
Collapse
Affiliation(s)
- Pedro M Martins
- Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.,IB-S - Institute for Research and Innovation on Bio-Sustainability, University of Minho, Braga 4710-057, Portugal
| | - Ana C Lima
- Centre/Department of Physics, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Sylvie Ribeiro
- Centre of Molecular and Environmental Biology (CBMA), Universidade do Minho, Campus de Gualtar, Braga 4710-057, Portugal.,Centre/Department of Physics, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| | - Senentxu Lanceros-Mendez
- 3BCMaterials, Basque Centre for Materials and Applications, UPV/EHU Science Park, Leioa 48940, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain
| | - Pedro Martins
- IB-S - Institute for Research and Innovation on Bio-Sustainability, University of Minho, Braga 4710-057, Portugal.,Centre/Department of Physics, University of Minho, Campus de Gualtar, Braga 4710-057, Portugal
| |
Collapse
|
38
|
Liu L, Ma Q, Cao J, Gao Y, Han S, Liang Y, Zhang T, Song Y, Sun Y. Recent progress of graphene oxide-based multifunctional nanomaterials for cancer treatment. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00087-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
In the last decade, graphene oxide-based nanomaterials, such as graphene oxide (GO) and reduced graphene oxide (rGO), have attracted more and more attention in the field of biomedicine. Due to the versatile surface functionalization, ultra-high surface area, and excellent biocompatibility of graphene oxide-based nanomaterials, which hold better promise for potential applications than among other nanomaterials in biomedical fields including drug/gene delivery, biomolecules detection, tissue engineering, especially in cancer treatment.
Results
Here, we review the recent progress of graphene oxide-based multifunctional nanomaterials for cancer treatment. A comprehensive and in-depth depiction of unique property of graphene oxide-based multifunctional nanomaterials is first interpreted, with particular descriptions about the suitability for applying in cancer therapy. Afterward, recently emerging representative applications of graphene oxide-based multifunctional nanomaterials in antitumor therapy, including as an ideal carrier for drugs/genes, phototherapy, and bioimaging, are systematically summarized. Then, the biosafety of the graphene oxide-based multifunctional nanomaterials is reviewed.
Conclusions
Finally, the conclusions and perspectives on further advancing the graphene oxide-based multifunctional nanomaterials toward potential and versatile development for fundamental researches and nanomedicine are proposed.
Graphic abstract
Collapse
|
39
|
Shrestha B, Wang L, Brey EM, Uribe GR, Tang L. Smart Nanoparticles for Chemo-Based Combinational Therapy. Pharmaceutics 2021; 13:853. [PMID: 34201333 PMCID: PMC8227511 DOI: 10.3390/pharmaceutics13060853] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/27/2022] Open
Abstract
Cancer is a heterogeneous and complex disease. Traditional cancer therapy is associated with low therapeutic index, acquired resistance, and various adverse effects. With the increasing understanding of cancer biology and technology advancements, more strategies have been exploited to optimize the therapeutic outcomes. The rapid development and application of nanomedicine have motivated this progress. Combinational regimen, for instance, has become an indispensable approach for effective cancer treatment, including the combination of chemotherapeutic agents, chemo-energy, chemo-gene, chemo-small molecules, and chemo-immunology. Additionally, smart nanoplatforms that respond to external stimuli (such as light, temperature, ultrasound, and magnetic field), and/or to internal stimuli (such as changes in pH, enzymes, hypoxia, and redox) have been extensively investigated to improve precision therapy. Smart nanoplatforms for combinational therapy have demonstrated the potential to be the next generation cancer treatment regimen. This review aims to highlight the recent advances in smart combinational therapy.
Collapse
Affiliation(s)
| | | | | | - Gabriela Romero Uribe
- Department of Biomedical and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (B.S.); (L.W.); (E.M.B.)
| | - Liang Tang
- Department of Biomedical and Chemical Engineering, The University of Texas at San Antonio, San Antonio, TX 78249, USA; (B.S.); (L.W.); (E.M.B.)
| |
Collapse
|
40
|
Khan FA, Albalawi R, Pottoo FH. Trends in targeted delivery of nanomaterials in colon cancer diagnosis and treatment. Med Res Rev 2021; 42:227-258. [PMID: 33891325 DOI: 10.1002/med.21809] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022]
Abstract
Colon cancer is an adenocarcinoma, which subsequently develops into malignant tumors, if not treated properly. The current colon cancer therapy mainly revolves around chemotherapy, radiotherapy and surgery, but the search continues for more effective interventions. With the advancement of nanoparticles (NPs), it is now possible to diagnose and treat colon cancers with different types, shapes, and sizes of NPs. Nanoformulations such as quantum dots, iron oxide, polymeric NPs, dendrimers, polypeptides, gold NPs, silver NPs, platinum NPs, and cerium oxide have been either extensively used alone or in combination with other nanomaterials or drugs in colon cancer diagnosis, and treatments. These nanoformulations possess high biocompatibility and bioavailability, which makes them the most suitable candidates for cancer treatment. The size and shape of NPs are critical to achieving an effective drug delivery in cancer treatment and diagnosis. Most NPs currently are under different testing phases (in vitro, preclinical, and clinical), whereas some of them have been approved for therapeutic applications. We have comprehensively reviewed the recent advances in the applications of NPs-based formulations in colon cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Firdos A Khan
- Department of Stem Cell Biology, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Reem Albalawi
- Department of Stem Cell Biology, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Student of the volunteer/training program at IRMC
| | - Faheem H Pottoo
- College of Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
41
|
Darwish MSA, Kim H, Bui MP, Le TA, Lee H, Ryu C, Lee JY, Yoon J. The Heating Efficiency and Imaging Performance of Magnesium Iron Oxide@tetramethyl Ammonium Hydroxide Nanoparticles for Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1096. [PMID: 33922608 PMCID: PMC8145217 DOI: 10.3390/nano11051096] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/05/2021] [Accepted: 04/16/2021] [Indexed: 12/30/2022]
Abstract
Multifunctional magnetic nanomaterials displaying high specific loss power (SLP) and high imaging sensitivity with good spatial resolution are highly desired in image-guided cancer therapy. Currently, commercial nanoparticles do not sufficiently provide such multifunctionality. For example, Resovist® has good image resolution but with a low SLP, whereas BNF® has a high SLP value with very low image resolution. In this study, hydrophilic magnesium iron oxide@tetramethyl ammonium hydroxide nanoparticles were prepared in two steps. First, hydrophobic magnesium iron oxide nanoparticles were fabricated using a thermal decomposition technique, followed by coating with tetramethyl ammonium hydroxide. The synthesized nanoparticles were characterized using XRD, DLS, TEM, zeta potential, UV-Vis spectroscopy, and VSM. The hyperthermia and imaging properties of the prepared nanoparticles were investigated and compared to the commercial nanoparticles. One-dimensional magnetic particle imaging indicated the good imaging resolution of our nanoparticles. Under the application of a magnetic field of frequency 614.4 kHz and strength 9.5 kA/m, nanoparticles generated heat with an SLP of 216.18 W/g, which is much higher than that of BNF (14 W/g). Thus, the prepared nanoparticles show promise as a novel dual-functional magnetic nanomaterial, enabling both high performance for hyperthermia and imaging functionality for diagnostic and therapeutic processes.
Collapse
Affiliation(s)
- Mohamed S. A. Darwish
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (M.S.A.D.); (H.K.); (M.P.B.); (T.-A.L.)
- Egyptian Petroleum Research Institute, 1 Ahmed El-Zomor Street, El Zohour Region, Nasr City, Cairo 11727, Egypt
| | - Hohyeon Kim
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (M.S.A.D.); (H.K.); (M.P.B.); (T.-A.L.)
| | - Minh Phu Bui
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (M.S.A.D.); (H.K.); (M.P.B.); (T.-A.L.)
| | - Tuan-Anh Le
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (M.S.A.D.); (H.K.); (M.P.B.); (T.-A.L.)
| | - Hwangjae Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea; (H.L.); (C.R.)
| | - Chiseon Ryu
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea; (H.L.); (C.R.)
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea; (H.L.); (C.R.)
| | - Jungwon Yoon
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju 61005, Korea; (M.S.A.D.); (H.K.); (M.P.B.); (T.-A.L.)
| |
Collapse
|
42
|
Abstract
In this review, we summarized recent advances in the development and biological applications of polymeric nanoparticles embedded with superparamagnetic iron oxide nanoparticles (SPIONs). Superparamagnetic polymeric nanoparticles include core-shell nanoparticles, superparamagnetic polymeric micelles and superparamagnetic polymersomes. They have potential for various biomedical applications, including magnetic resonance imaging (MRI) contrast agents, drug delivery, detection of bacteria, viruses and proteins, etc. Finally, the challenges in the design and preparation of superparamagnetic nanoparticles towards clinical applications are explored and the prospects in this field are proposed.
Collapse
Affiliation(s)
- Yufen Xiao
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China.
| | | |
Collapse
|
43
|
Mohapatra A, Uthaman S, Park IK. External and Internal Stimuli-Responsive Metallic Nanotherapeutics for Enhanced Anticancer Therapy. Front Mol Biosci 2021; 7:597634. [PMID: 33505987 PMCID: PMC7831291 DOI: 10.3389/fmolb.2020.597634] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Therapeutic, diagnostic, and imaging approaches based on nanotechnology offer distinct advantages in cancer treatment. Various nanotherapeutics have been presented as potential alternatives to traditional anticancer therapies such as chemotherapy, radiotherapy, and surgical intervention. Notably, the advantage of nanotherapeutics is mainly attributable to their accumulation and targeting ability toward cancer cells, multiple drug-carrying abilities, combined therapies, and imaging approaches. To date, numerous nanoparticle formulations have been developed for anticancer therapy and among them, metallic nanotherapeutics reportedly demonstrate promising cancer therapeutic and diagnostic efficiencies owing to their dense surface functionalization ability, uniform size distribution, and shape-dependent optical responses, easy and cost-effective synthesis procedure, and multiple anti-cancer effects. Metallic nanotherapeutics can remodel the tumor microenvironment by changing unfavorable therapeutic conditions into therapeutically accessible ones with the help of different stimuli, including light, heat, ultrasound, an alternative magnetic field, redox, and reactive oxygen species. The combination of metallic nanotherapeutics with both external and internal stimuli can be used to trigger the on-demand release of therapeutic molecules, augmenting the therapeutic efficacies of anticancer therapies such as photothermal therapy, photodynamic therapy, magnetic hyperthermia, sonodynamic therapy, chemodynamic therapy, and immunotherapy. In this review, we have summarized the role of different metallic nanotherapeutics in anti-cancer therapy, as well as their combinational effects with multiple stimuli for enhanced anticancer therapy.
Collapse
Affiliation(s)
- Adityanarayan Mohapatra
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeollanam-do, South Korea
| | - Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon, South Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, Jeollanam-do, South Korea
| |
Collapse
|
44
|
Wang W, Huang Z, Huang Y, Pan X, Wu C. Updates on the applications of iron-based nanoplatforms in tumor theranostics. Int J Pharm 2020; 589:119815. [PMID: 32877726 DOI: 10.1016/j.ijpharm.2020.119815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/28/2020] [Accepted: 08/23/2020] [Indexed: 12/11/2022]
Abstract
With the development of biomedicine and materials science, the emerging research of iron-based nanoplatforms (INPs) have provided a bright future for tumor theranostics. Thanks to its excellent biocompatibility and diverse application potential, some INPs have successfully transformed from the laboratory to the clinic and market, making it one of the most successful nanoplatforms. Further investigations associated with its enormous biomedical potential is continuing, and new features of them are being demonstrated. The discovery of ferroptosis therapy opens up new avenue for the applications of INPs in tumor therapy, which is attracting tremendous attention from worldwide. It is well established that some of the INPs are capable of triggering the tumor cell ferroptosis efficiently, accelerating the tumor cell death process. Combined with anti-tumor drugs or other tumor therapy approaches, the INPs-induced ferroptosis are expected to break the bottleneck in the treatment of drug-resistant malignant tumors. In addition, other applications of INPs in tumor theranostics field are still active. Featured with the catalase-like ability, INPs were also well documented to reverse the tumor hypoxia as nanozymes, assisting and enhancing the oxygen-consuming tumor therapy approaches. And the unique magnetic property of INPs endow it with great potential in tumor diagnosis, hyperthermal therapy and target drug delivery. It is of great significance to summarize these new advances. Herein, the latest reports of the applications of INPs in tumor theranostics are classified to expound the trend of its research and development. The featured functions of it will be discussed in detail to provide a new insight. The key issues needing to be addressed and the development prospective will be put forward. We hope that this review will be helpful to understand the ample potential of INPs in tumor theranostics field.
Collapse
Affiliation(s)
- Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China.
| | - Zhengwei Huang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China.
| | - Ying Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, Guangdong, PR China.
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China.
| | - Chuanbin Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China.
| |
Collapse
|
45
|
Kruszewska J, Sikorski J, Samsonowicz-Górski J, Matczuk M. A CE-ICP-MS/MS method for the determination of superparamagnetic iron oxide nanoparticles under simulated physiological conditions. Anal Bioanal Chem 2020; 412:8145-8153. [PMID: 32968852 PMCID: PMC7584539 DOI: 10.1007/s00216-020-02948-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 11/24/2022]
Abstract
Over the past few years, superparamagnetic iron oxide nanoparticles (SPIONs) have attracted much attention due to their medicinally attractive properties and their possible application in cancer diagnosis and therapy. However, there is still a lack of appropriate methods to enable quantitative monitoring of the particle changes in a physiological environment, which could be beneficial for evaluating their in vitro and in vivo behavior. For this reason, the main goal of this study was the development of a novel capillary electrophoresis-inductively coupled plasma mass spectrometry (CE-ICP-MS/MS) method for the determination of SPIONs suitable for the future examination of their changes upon incubation with proteins under simulated physiological conditions. The type and flow rate of the collision/reaction gas were chosen with the aim of simultaneous monitoring of Fe and S. The type and concentration of the background electrolyte, applied voltage, and sample loading were optimized to obtain SPION signals of the highest intensity and minimum half-width of the peak. Analytical parameters were at a satisfactory level: reproducibility (intra- and inter-day) of migration times and peak areas (presented as RSD) in the range of 0.23-4.98%, recovery: 96.7% and 93.3%, the limit of detection (for monitoring 56Fe16O+ by mass-shift approach) 54 ng mL-1 Fe (0.97 μM) and 101 ng mL-1 Fe (1.82 μM) for SPIONs with carboxyl and amino terminal groups, respectively. To the best of our knowledge, this is the first reported use of CE-ICP-MS/MS for the quantification of SPIONs and monitoring of interactions with proteins.
Collapse
Affiliation(s)
- Joanna Kruszewska
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664, Warsaw, Poland
| | - Jacek Sikorski
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664, Warsaw, Poland
| | - Jan Samsonowicz-Górski
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664, Warsaw, Poland
| | - Magdalena Matczuk
- Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664, Warsaw, Poland.
| |
Collapse
|
46
|
Etemadi H, Plieger PG. Magnetic Fluid Hyperthermia Based on Magnetic Nanoparticles: Physical Characteristics, Historical Perspective, Clinical Trials, Technological Challenges, and Recent Advances. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000061] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hossein Etemadi
- School of Fundamental Sciences Massey University Palmerston North 4474 New Zealand
| | - Paul G. Plieger
- School of Fundamental Sciences Massey University Palmerston North 4474 New Zealand
| |
Collapse
|
47
|
Hamza H, Ferretti AM, Innocenti C, Fidecka K, Licandro E, Sangregorio C, Maggioni D. An Approach for Magnetic Halloysite Nanocomposite with Selective Loading of Superparamagnetic Magnetite Nanoparticles in the Lumen. Inorg Chem 2020; 59:12086-12096. [PMID: 32805986 PMCID: PMC8009513 DOI: 10.1021/acs.inorgchem.0c01039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
We present for the
first time a method for the preparation of magnetic
halloysite nanotubes (HNT) by loading of preformed superparamagnetic
magnetite nanoparticles (SPION) of diameter size ∼6 nm with
a hydrodynamic diameter of ∼10 nm into HNT. We found that the
most effective route to reach this goal relies on the modification
of the inner lumen of HNT by tetradecylphosphonic acid (TDP) to give
HNT–TDP, followed by the loading with preformed oleic acid
(OA)-stabilized SPION. Transmission electron microscopy evidenced
the presence of highly crystalline magnetic nanoparticles only in
the lumen, partially ordered in chainlike structures. Conversely,
attempts to obtain the same result by exploiting either the positive
charge of the HNT inner lumen employing SPIONs covered with negatively
charged capping agents or the in situ synthesis of
SPION by thermal decomposition were not effective. HNT–TDP
were characterized by infrared spectroscopy (ATR-FTIR), thermogravimetric
analysis (TGA), and ζ-potential, and all of the techniques confirmed
the presence of TDP onto the HNT. Moreover, the inner localization
of TDP was ascertained by the use of Nile Red, a molecule whose luminescence
is very sensitive to the polarity of the environment. The free SPION@OA
(as a colloidal suspension and as a powder) and SPION-in-HNT powder
were magnetically characterized by measuring the ZFC-FC magnetization
curves as well as the hysteresis cycles at 300 and 2.5 K, confirming
that the super-paramagnetic behavior and the main magnetic properties
of the free SPION were preserved once embedded in SPION-in-HNT. SPION nanoparticles are selectively loaded
into halloysite
lumen, keeping their superparamagnetic character.
Collapse
Affiliation(s)
- Hady Hamza
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | | | - Claudia Innocenti
- ICCOM-CNR, via Madonna del Piano 10, 50019 Sesto, Fiorentino, Italy.,Consorzio INSTM, Via G. Giusti, 9, 50121 Firenze, Italy.,Dipartimento di Chimica, Università degli Studi di Firenze, via della Lastruccia 3, 50019 Sesto, Fiorentino, Italy
| | - Katarzyna Fidecka
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Emanuela Licandro
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy
| | - Claudio Sangregorio
- ICCOM-CNR, via Madonna del Piano 10, 50019 Sesto, Fiorentino, Italy.,Consorzio INSTM, Via G. Giusti, 9, 50121 Firenze, Italy.,Dipartimento di Chimica, Università degli Studi di Firenze, via della Lastruccia 3, 50019 Sesto, Fiorentino, Italy
| | - Daniela Maggioni
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133 Milano, Italy.,Consorzio INSTM, Via G. Giusti, 9, 50121 Firenze, Italy
| |
Collapse
|
48
|
Gauger AJ, Hershberger KK, Bronstein LM. Theranostics Based on Magnetic Nanoparticles and Polymers: Intelligent Design for Efficient Diagnostics and Therapy. Front Chem 2020; 8:561. [PMID: 32733850 PMCID: PMC7359411 DOI: 10.3389/fchem.2020.00561] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/02/2020] [Indexed: 12/21/2022] Open
Abstract
Theranostics is a fast-growing field due to demands for new, efficient therapeutics which could be precisely delivered to the target site using multimodal imaging with enhancing auxiliary actions. In this review article we discuss theranostic nanoplatforms containing polymers and magnetic nanoparticles along with other components. Magnetic nanoparticles allow for both diagnostic and therapeutic (hyperthermia) capabilities, while polymers can be reservoirs for drugs and are easily functionalized for cell targeting. We focus on the most important design strategies to achieve optimal theranostic effects as well as the roles of different components included in theranostics, reviewing the literature from the last 5 years.
Collapse
Affiliation(s)
- Andrew J. Gauger
- Department of Chemistry, Indiana University, Bloomington, IN, United States
| | | | - Lyudmila M. Bronstein
- Department of Chemistry, Indiana University, Bloomington, IN, United States
- Department of Physics, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
49
|
Pham SH, Choi Y, Choi J. Stimuli-Responsive Nanomaterials for Application in Antitumor Therapy and Drug Delivery. Pharmaceutics 2020; 12:E630. [PMID: 32635539 PMCID: PMC7408499 DOI: 10.3390/pharmaceutics12070630] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 01/14/2023] Open
Abstract
The new era of nanotechnology has produced advanced nanomaterials applicable to various fields of medicine, including diagnostic bio-imaging, chemotherapy, targeted drug delivery, and biosensors. Various materials are formed into nanoparticles, such as gold nanomaterials, carbon quantum dots, and liposomes. The nanomaterials have been functionalized and widely used because they are biocompatible and easy to design and prepare. This review mainly focuses on nanomaterials responsive to the external stimuli used in drug-delivery systems. To overcome the drawbacks of conventional therapeutics to a tumor, the dual- and multi-responsive behaviors of nanoparticles have been harnessed to improve efficiency from a drug delivery point of view. Issues and future research related to these nanomaterial-based stimuli sensitivities and the scope of stimuli-responsive systems for nanomedicine applications are discussed.
Collapse
Affiliation(s)
| | | | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Korea; (S.H.P.); (Y.C.)
| |
Collapse
|
50
|
Lim ZW, Varma VB, Ramanujan RV, Miserez A. Magnetically responsive peptide coacervates for dual hyperthermia and chemotherapy treatments of liver cancer. Acta Biomater 2020; 110:221-230. [PMID: 32422317 DOI: 10.1016/j.actbio.2020.04.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/08/2020] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
Liver cancer is an aggressive malignancy associated with high levels of mortality and morbidity. Doxorubicin (Dox) is often used to slow down liver cancer progression; however its efficacy is limited, and its severe side effects prevent its routine use at therapeutic concentrations. We present a biomimetic peptide that coacervates into micro-droplets, within which both Dox and magnetic nanoparticles (MNPs) can be sequestered. These Dox-loaded Magnetic Coacervates (DMCs) can be used for thermo-chemotherapy, with the controlled release of Dox triggered by an external Alternating Magnetic Field (AMF). The DMCs are internalized by the cells via an energy-independent mechanism which is not based on endocytosis. Application of AMF generates a temperature of 45 °C within the DMCs, triggering their disassembly and the simultaneous release of Dox, thereby resulting in dual hyperthermia and chemotherapy for more efficient cancer therapy. In vitro studies conducted under AMF reveal that DMCs are cytocompatible and effective in inducing HepG2 liver cancer cell death. Thermo-chemotherapy treatment against HepG2 cells is also shown to be more effective compared to either hyperthermia or chemotherapy treatments alone. Thus, our novel peptide DMCs can open avenues in theranostic strategies against liver cancer through programmable, wireless, and remote control of Dox release. STATEMENT OF SIGNIFICANCE: Simultaneous administration of chemical and thermal therapy (thermo-chemotherapy) is more effective in inducing liver cancer cell death and improving survival rate. Thus, there is a keen interest in developing suitable carriers for thermo-chemotherapy. Coacervate micro-droplets display significant advantages, including high loading capacity, fast self-assembly in aqueous environments, and liquid-like behavior. However, they have not yet been explored as carriers for thermo-chemotherapy. Here, we demonstrate that peptide coacervate micro-droplets can co-encapsulate Dox and magnetic nanoparticles and cross the cell membrane. Applying an alternating magnetic field to cells containing drug-loaded coacervates triggers the release of Dox as well as the localized heating by magnetic hyperthermia, resulting in efficient liver cancer cell death by dual thermo-chemotherapy.
Collapse
Affiliation(s)
- Zhi Wei Lim
- Biological and Biomimetic Materials Laboratory, Centre for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 637553
| | - Vijaykumar B Varma
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798
| | - Raju V Ramanujan
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798
| | - Ali Miserez
- Biological and Biomimetic Materials Laboratory, Centre for Biomimetic Sensor Science, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 637553; School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551.
| |
Collapse
|