1
|
Luo L, Zhou H, Wang S, Pang M, Zhang J, Hu Y, You J. The Application of Nanoparticle-Based Imaging and Phototherapy for Female Reproductive Organs Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2207694. [PMID: 37154216 DOI: 10.1002/smll.202207694] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/06/2023] [Indexed: 05/10/2023]
Abstract
Various female reproductive disorders affect millions of women worldwide and bring many troubles to women's daily life. Let alone, gynecological cancer (such as ovarian cancer and cervical cancer) is a severe threat to most women's lives. Endometriosis, pelvic inflammatory disease, and other chronic diseases-induced pain have significantly harmed women's physical and mental health. Despite recent advances in the female reproductive field, the existing challenges are still enormous such as personalization of disease, difficulty in diagnosing early cancers, antibiotic resistance in infectious diseases, etc. To confront such challenges, nanoparticle-based imaging tools and phototherapies that offer minimally invasive detection and treatment of reproductive tract-associated pathologies are indispensable and innovative. Of late, several clinical trials have also been conducted using nanoparticles for the early detection of female reproductive tract infections and cancers, targeted drug delivery, and cellular therapeutics. However, these nanoparticle trials are still nascent due to the body's delicate and complex female reproductive system. The present review comprehensively focuses on emerging nanoparticle-based imaging and phototherapies applications, which hold enormous promise for improved early diagnosis and effective treatments of various female reproductive organ diseases.
Collapse
Affiliation(s)
- Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, P. R. China
| | - Huanli Zhou
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, P. R. China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, P. R. China
| | - Mei Pang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, P. R. China
| | - Junlei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, P. R. China
| | - Yilong Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, P. R. China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, P. R. China
| |
Collapse
|
2
|
Tian Y, He X, Yuan Y, Zhang S, Wang C, Dong J, Liu Z, Jing H. TME-Responsive Nanoplatform with Glutathione Depletion for Enhanced Tumor-Specific Mild Photothermal/Gene/Ferroptosis Synergistic Therapy. Int J Nanomedicine 2024; 19:9145-9160. [PMID: 39258005 PMCID: PMC11386068 DOI: 10.2147/ijn.s475698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024] Open
Abstract
Background Triple negative breast cancer (TNBC) is one of the worst prognosis types of breast cancer that urgently needs effective therapy methods. However, cancer is a complicated disease that usually requires multiple treatment modalities. Methods A tumor microenvironment (TME)-responsive PFC/TRIM37@Fe-TA@HA (abbreviated as PTFTH) nanoplatform was constructed by coating Fe3+ and tannic acid (TA) on the surface of TRIM37-siRNA loaded phase-transition perfluorocarbon (PFC) nanodroplets and further modifying them with hyaluronic acid (HA) to achieve tumor-specific mild photothermal/gene/ferroptosis synergistic therapy (MPTT/GT/ Ferroptosis) in vitro. Once internalized into tumor cells through CD44 receptor-mediated active targeting, the HA shell of PTFTH would be preliminarily disassembled by hyaluronidase (HAase) to expose the Fe-TA metal-phenolic networks (MPNs), which would further degrade in response to an acidic lysosomal environment, leading to HAase/pH dual-responsive release of Fe3+ and PFC/TRIM37. Results PTFTH showed good biocompatibility in vitro. On the one hand, the released Fe3+ could deplete the overexpressed glutathione (GSH) through redox reactions and produce Fe2+, which in turn converts endogenous H2O2 into highly cytotoxic hydroxyl radicals (•OH) for chemodynamic therapy (CDT). On the other hand, the local hyperthermia generated by PTFTH under 808 nm laser irradiation could not only improve CDT efficacy through accelerating the Fe2+-mediated Fenton reaction, but also enhance TRIM37-siRNA delivery for gene therapy (GT). The consumption of GSH and accumulation of •OH synergistically augmented intracellular oxidative stress, resulting in substantial tumor cell ferroptosis. Moreover, PTFTH possessed outstanding contrast enhanced ultrasound (CEUS), photoacoustic imaging (PAI) and magnetic resonance imaging (MRI) ability. Conclusion This PTFTH based multiple-mode therapeutic strategy has successfully achieved a synergistic anticancer effect in vitro and has the potential to be translated into clinical application for tumor therapy in future.
Collapse
Affiliation(s)
- Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Shijie Zhang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Jialin Dong
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, 150081, People's Republic of China
| |
Collapse
|
3
|
Hheidari A, Mohammadi J, Ghodousi M, Mahmoodi M, Ebrahimi S, Pishbin E, Rahdar A. Metal-based nanoparticle in cancer treatment: lessons learned and challenges. Front Bioeng Biotechnol 2024; 12:1436297. [PMID: 39055339 PMCID: PMC11269265 DOI: 10.3389/fbioe.2024.1436297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Cancer, being one of the deadliest diseases, poses significant challenges despite the existence of traditional treatment approaches. This has led to a growing demand for innovative pharmaceutical agents that specifically target cancer cells for effective treatment. In recent years, the use of metal nanoparticles (NPs) as a promising alternative to conventional therapies has gained prominence in cancer research. Metal NPs exhibit unique properties that hold tremendous potential for various applications in cancer treatment. Studies have demonstrated that certain metals possess inherent or acquired anticancer capabilities through their surfaces. These properties make metal NPs an attractive focus for therapeutic development. In this review, we will investigate the applicability of several distinct classes of metal NPs for tumor targeting in cancer treatment. These classes may include gold, silver, iron oxide, and other metals with unique properties that can be exploited for therapeutic purposes. Additionally, we will provide a comprehensive summary of the risk factors associated with the therapeutic application of metal NPs. Understanding and addressing these factors will be crucial for successful clinical translation and to mitigate any potential challenges or failures in the translation of metal NP-based therapies. By exploring the therapeutic potential of metal NPs and identifying the associated risk factors, this review aims to contribute to the advancement of cancer treatment strategies. The anticipated outcome of this review is to provide valuable insights and pave the way for the advancement of effective and targeted therapies utilizing metal NPs specifically for cancer patients.
Collapse
Affiliation(s)
- Ali Hheidari
- Department of Mechanical Engineering, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Javad Mohammadi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Maryam Ghodousi
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, United States
| | - Mohammadreza Mahmoodi
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Sina Ebrahimi
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Esmail Pishbin
- Bio-microfluidics Lab, Department of Electrical Engineering and Information Technology, Iranian Research Organization for Science and Technology, Tehran, Iran
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran
| |
Collapse
|
4
|
Su J, Mao X, Wang L, Chen Z, Wang W, Zhao C, Li G, Guo W, Hu Y. Lactate/GPR81 recruits regulatory T cells by modulating CX3CL1 to promote immune resistance in a highly glycolytic gastric cancer. Oncoimmunology 2024; 13:2320951. [PMID: 38419759 PMCID: PMC10900271 DOI: 10.1080/2162402x.2024.2320951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Lactate plays an important role in shaping immune tolerance in tumor microenvironment (TME) and correlates with poor prognosis in various solid tumors. Overcoming the immune resistance in an acidic TME may improve the anti-tumor immunity. Here, this study elucidated that via G-protein-coupled receptor 81 (GPR81), lactate could modulate immune tolerance in TME by recruiting regulatory T cells (Tregs) in vitro and in vivo. A high concentration of lactate was detected in cell supernatant and tissues of gastric cancer (GC), which was modulated by lactic dehydrogenase A (LDHA). GPR81 was the natural receptor of lactate and was overexpressed in different GC cell lines and samples, which correlated with poor outcomes in GC patients. Lactate/GPR81 signaling could promote the infiltration of Tregs into TME by inducing the expression of chemokine CX3CL1. GPR81 deficiency could decrease the infiltration of Tregs into TME, thereby inhibiting GC progression by weakening the inhibition of CD8+T cell function in a humanized mouse model. In conclusion, targeting the lactate/GPR81 signaling may potentially serve as a critical process to overcome immune resistance in highly glycolytic GC.
Collapse
Affiliation(s)
- Jin Su
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of General Surgery, Zhuzhou Hospital affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Xinyuan Mao
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Lingzhi Wang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhian Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weisheng Wang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Cuiyin Zhao
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Guoxin Li
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weihong Guo
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanfeng Hu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Ruhoff V, Arastoo MR, Moreno-Pescador G, Bendix PM. Biological Applications of Thermoplasmonics. NANO LETTERS 2024; 24:777-789. [PMID: 38183300 PMCID: PMC10811673 DOI: 10.1021/acs.nanolett.3c03548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/08/2024]
Abstract
Thermoplasmonics has emerged as an extraordinarily versatile tool with profound applications across various biological domains ranging from medical science to cell biology and biophysics. The key feature of nanoscale plasmonic heating involves remote activation of heating by applying laser irradiation to plasmonic nanostructures that are designed to optimally convert light into heat. This unique capability paves the way for a diverse array of applications, facilitating the exploration of critical biological processes such as cell differentiation, repair, signaling, and protein functionality, and the advancement of biosensing techniques. Of particular significance is the rapid heat cycling that can be achieved through thermoplasmonics, which has ushered in remarkable technical innovations such as accelerated amplification of DNA through quantitative reverse transcription polymerase chain reaction. Finally, medical applications of photothermal therapy have recently completed clinical trials with remarkable results in prostate cancer, which will inevitably lead to the implementation of photothermal therapy for a number of diseases in the future. Within this review, we offer a survey of the latest advancements in the burgeoning field of thermoplasmonics, with a keen emphasis on its transformative applications within the realm of biosciences.
Collapse
Affiliation(s)
| | - Mohammad Reza Arastoo
- Niels
Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
| | - Guillermo Moreno-Pescador
- Niels
Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
- Copenhagen
Plant Science Center, Department of Plant and Environmental Sciences, University of Copenhagen, Thorvaldsensvej 40, 1871 Frederiksberg, Denmark
| | - Poul Martin Bendix
- Niels
Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100 København Ø, Denmark
| |
Collapse
|
6
|
Batool S, Sohail S, Ud Din F, Alamri AH, Alqahtani AS, Alshahrani MA, Alshehri MA, Choi HG. A detailed insight of the tumor targeting using nanocarrier drug delivery system. Drug Deliv 2023; 30:2183815. [PMID: 36866455 DOI: 10.1080/10717544.2023.2183815] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023] Open
Abstract
Human struggle against the deadly disease conditions is continued since ages. The contribution of science and technology in fighting against these diseases cannot be ignored exclusively due to the invention of novel procedure and products, extending their size ranges from micro to nano. Recently nanotechnology has been gaining more consideration for its ability to diagnose and treat different cancers. Different nanoparticles have been used to evade the issues related with conservative anticancer delivery systems, including their nonspecificity, adverse effects and burst release. These nanocarriers including, solid lipid nanoparticles (SLNs), liposomes, nano lipid carriers (NLCs), nano micelles, nanocomposites, polymeric and magnetic nanocarriers, have brought revolutions in antitumor drug delivery. Nanocarriers improved the therapeutic efficacy of anticancer drugs with better accumulation at the specific site with sustained release, improved bioavailability and apoptosis of the cancer cells while bypassing the normal cells. In this review, the cancer targeting techniques and surface modification on nanoparticles are discussed briefly with possible challenges and opportunities. It can be concluded that understanding the role of nanomedicine in tumor treatment is significant, and therefore, the modern progressions in this arena is essential to be considered for a prosperous today and an affluent future of tumor patients.
Collapse
Affiliation(s)
- Sibgha Batool
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saba Sohail
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Fakhar Ud Din
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.,Nanomedicine Research Group, Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ali H Alamri
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ahmad S Alqahtani
- Department of Pharmacy, Mental Health Hospital, Ministry of Health, Abha, Saudi Arabia
| | - Mohammad A Alshahrani
- Department of Medical Supply in Khamis Mushet General Hospital, Ministry of Health, Khamis Mushet, Saudi Arabia
| | - Mohammed A Alshehri
- Department of Pharmacy, Abha Maternity and Children Hospital, Ministry of Health, Abha, Saudi Arabia
| | - Han Gon Choi
- College of Pharmacy & Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, South Korea
| |
Collapse
|
7
|
Jiang L, Qi Y, Yang L, Miao Y, Ren W, Liu H, Huang Y, Huang S, Chen S, Shi Y, Cai L. Remodeling the tumor immune microenvironment via siRNA therapy for precision cancer treatment. Asian J Pharm Sci 2023; 18:100852. [PMID: 37920650 PMCID: PMC10618707 DOI: 10.1016/j.ajps.2023.100852] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/19/2023] [Accepted: 08/02/2023] [Indexed: 11/04/2023] Open
Abstract
How to effectively transform the pro-oncogenic tumor microenvironments (TME) surrounding a tumor into an anti-tumoral never fails to attract people to study. Small interfering RNA (siRNA) is considered one of the most noteworthy research directions that can regulate gene expression following a process known as RNA interference (RNAi). The research about siRNA delivery targeting tumor cells and TME has been on the rise in recent years. Using siRNA drugs to silence critical proteins in TME was one of the most efficient solutions. However, the manufacture of a siRNA delivery system faces three major obstacles, i.e., appropriate cargo protection, accurately targeted delivery, and site-specific cargo release. In the following review, we summarized the pharmacological actions of siRNA drugs in remolding TME. In addition, the delivery strategies of siRNA drugs and combination therapy with siRNA drugs to remodel TME are thoroughly discussed. In the meanwhile, the most recent advancements in the development of all clinically investigated and commercialized siRNA delivery technologies are also presented. Ultimately, we propose that nanoparticle drug delivery siRNA may be the future research focus of oncogene therapy. This summary offers a thorough analysis and roadmap for general readers working in the field.
Collapse
Affiliation(s)
- Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yao Qi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lei Yang
- Department of Pharmacy, Jianyang People's Hospital of Sichuan Province, Jianyang 641400, China
| | - Yangbao Miao
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Weiming Ren
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Hongmei Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yi Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Shan Huang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Shiyin Chen
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Lulu Cai
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| |
Collapse
|
8
|
Role of Tunable Gold Nanostructures in Cancer Nanotheranostics: Implications on Synthesis, Toxicity, Clinical Applications and Their Associated Opportunities and Challenges. JOURNAL OF NANOTHERANOSTICS 2023. [DOI: 10.3390/jnt4010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The existing diagnosis and treatment modalities have major limitations related to their precision and capability to understand several stages of disease development. A superior therapeutic system consists of a multifunctional approach in early diagnosis of the disease with a simultaneous progressive cure, using a precise medical approach towards complex treatment. These challenges can be addressed via nanotheranostics and explore suitable approaches to improve health care. Nanotechnology in combination with theranostics as an unconventional platform paved the way for developing novel strategies and modalities leading to diagnosis and therapy for complex disease conditions, ranging from acute to chronic levels. Among the metal nanoparticles, gold nanoparticles are being widely used for theranostics due to their inherent non-toxic nature and plasmonic properties. The unique optical and chemical properties of plasmonic metal nanoparticles along with theranostics have led to a promising era of plausible early detection of disease conditions, and they enable real-time monitoring with enhanced non-invasive or minimally invasive imaging of several ailments. This review aims to highlight the improvement and advancement brought to nanotheranostics by gold nanoparticles in the past decade. The clinical use of the metal nanoparticles in nanotheranostics is explained, along with the future perspectives on addressing the key applications related to diagnostics and therapeutics, respectively. The scope of gold nanoparticles and their realistic potential to design a sophisticated theranostic system is discussed in detail, along with their implications in clinical advancements which are the needs of the hour. The review concluded with the challenges, opportunities, and implications on translational potential of using gold nanoparticles in nanotheranostics.
Collapse
|
9
|
A rationally designed cancer vaccine based on NIR-II fluorescence image-guided light-triggered remote control of antigen cross-presentation and autophagy. Acta Pharm Sin B 2022. [PMID: 37521873 PMCID: PMC10373097 DOI: 10.1016/j.apsb.2022.11.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Cancer vaccines represent a promising immunotherapeutic treatment modality. The promotion of cross-presentation of extracellular tumor-associated antigens on the major histocompatibility complex (MHC) class I molecules and dendritic cell maturation at the appropriate time and place is crucial for cancer vaccines to prime cytolytic T cell response with reduced side effects. Current vaccination strategies, however, are not able to achieve the spatiotemporal control of antigen cross-presentation. Here, we report a liposomal vaccine loading the second near-infrared window (NIR-II, 1000-1700 nm) fluorophore BPBBT with an efficient photothermal conversion effect that offers an NIR-light-triggered endolysosomal escape under the imaging guidance. The NIR-II image-guided vaccination strategy specifically controls the cytosolic delivery of antigens for cross-presentation in the draining lymph nodes (DLNs). Moreover, the photothermally induced endolysosomal rupture initiates autophagy. We also find that the adjuvant simvastatin acts as an autophagy activator through inhibiting the PI3K/AKT/mTOR pathway. The light-induced autophagy in the DLNs together with simvastatin treatment cooperatively increase MHC class II expression by activating autophagy machinery for dendritic cell maturation. This study presents a paradigm of NIR-II image-guided light-triggered vaccination. The approach for remote control of antigen cross-presentation and autophagy represents a new strategy for vaccine development.
Collapse
|
10
|
Farjadian F, Ghasemi S, Akbarian M, Hoseini-Ghahfarokhi M, Moghoofei M, Doroudian M. Physically stimulus-responsive nanoparticles for therapy and diagnosis. Front Chem 2022; 10:952675. [PMID: 36186605 PMCID: PMC9515617 DOI: 10.3389/fchem.2022.952675] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Nanoparticles offer numerous advantages in various fields of science, particularly in medicine. Over recent years, the use of nanoparticles in disease diagnosis and treatments has increased dramatically by the development of stimuli-responsive nano-systems, which can respond to internal or external stimuli. In the last 10 years, many preclinical studies were performed on physically triggered nano-systems to develop and optimize stable, precise, and selective therapeutic or diagnostic agents. In this regard, the systems must meet the requirements of efficacy, toxicity, pharmacokinetics, and safety before clinical investigation. Several undesired aspects need to be addressed to successfully translate these physical stimuli-responsive nano-systems, as biomaterials, into clinical practice. These have to be commonly taken into account when developing physically triggered systems; thus, also applicable for nano-systems based on nanomaterials. This review focuses on physically triggered nano-systems (PTNSs), with diagnostic or therapeutic and theranostic applications. Several types of physically triggered nano-systems based on polymeric micelles and hydrogels, mesoporous silica, and magnets are reviewed and discussed in various aspects.
Collapse
Affiliation(s)
- Fatemeh Farjadian
- Pharmaceutical Sciences Research Center, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
- *Correspondence: Fatemeh Farjadian, , Soheila Ghasemi, , Mohammad Doroudian,
| | - Soheila Ghasemi
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran
- *Correspondence: Fatemeh Farjadian, , Soheila Ghasemi, , Mohammad Doroudian,
| | - Mohsen Akbarian
- Department of Chemistry, National Cheng Kung University, Tainan, Taiwan
| | | | - Mohsen Moghoofei
- Department of Microbiology, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
- *Correspondence: Fatemeh Farjadian, , Soheila Ghasemi, , Mohammad Doroudian,
| |
Collapse
|
11
|
Valencia FJ, Ramírez M, Varas A, Rogan J. Thermal Sensitivity on Eccentric Gold Hollow Nanoparticles: A Perspective from Atomistic Simulations. J Chem Inf Model 2021; 61:5499-5507. [PMID: 34726404 DOI: 10.1021/acs.jcim.1c00849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Eccentricity is a common feature consequence of several synthesis protocols of hollow nanoshells. Despite the crescent interest in these nanoparticles, it is still unclear how an irregular layer on the nanoparticle impacts the macroscopic properties. Here, we study the thermal stability of eccentric hollow nanoparticles (hNPs) for different sizes and eccentricity values by means of classical molecular dynamics simulations. Our results reveal that eccentricity displays a significant role in the thermal stability of hNPs. We attribute this behavior to the irregular shell contour, which collapses due to the thermal-activated diffusive process from the nanoparticle shell's most thin region. The mechanism is driven at low temperature by the nucleation of stacking faults until the amorphization for larger temperature values. Besides, for some particular eccentric hNPs, the shell suffers a surface reconstruction process, transforming the eccentric hNP into a concentric hNP. We believe that our study on thermal effects in eccentric hNPs has relevance because of their outstanding applications for plasmonic and sensing.
Collapse
Affiliation(s)
- Felipe J Valencia
- Centro de Investigación DAiTA Lab, Facultad de Estudios Interdisciplinarios, Universidad Mayor, Santiago 7510041, Chile.,Centro para el Desarrollo de la Nanociencia y la Nanotecnología, CEDENNA, Avda. Ecuador 3493, Santiago 9170124, Chile
| | - Max Ramírez
- Departamento de Física, Facultad de Ciencias, Universidad de Chile, Casilla 653, Santiago 7800024, Chile.,Centro para el Desarrollo de la Nanociencia y la Nanotecnologí a, CEDENNA, Avda. Ecuador 3493, Santiago 9170124, Chile
| | - Alejandro Varas
- Departamento de Física, Facultad de Ciencias, Universidad de Chile, Casilla 653, Santiago 7800024, Chile.,Centro para el Desarrollo de la Nanociencia y la Nanotecnologí a, CEDENNA, Avda. Ecuador 3493, Santiago 9170124, Chile
| | - José Rogan
- Departamento de Física, Facultad de Ciencias, Universidad de Chile, Casilla 653, Santiago 7800024, Chile.,Centro para el Desarrollo de la Nanociencia y la Nanotecnologí a, CEDENNA, Avda. Ecuador 3493, Santiago 9170124, Chile
| |
Collapse
|
12
|
Inbaraj BS, Hua LH, Chen BH. Comparative Study on Inhibition of Pancreatic Cancer Cells by Resveratrol Gold Nanoparticles and a Resveratrol Nanoemulsion Prepared from Grape Skin. Pharmaceutics 2021; 13:pharmaceutics13111871. [PMID: 34834286 PMCID: PMC8622665 DOI: 10.3390/pharmaceutics13111871] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/19/2021] [Accepted: 10/29/2021] [Indexed: 01/04/2023] Open
Abstract
Resveratrol, a phenolic compound possessing vital biological activities such as anti-cancer, is present abundantly in grape skin, a waste produced during the processing of grape juice. The objectives of this study were to prepare resveratrol-gold nanoparticles and a resveratrol nanoemulsion from grape skin and study their inhibition effects on pancreatic cancer cells BxPC-3. The spherical-shaped citrate gold nanoparticles (GNPs) and resveratrol-gold nanoparticles (R-GNPs) were, respectively, prepared with a surface plasmon resonance peak at 528 and 538 nm, mean particle size of 20.8 and 11.9 nm, and zeta-potential at −32.7 and −66.7 mV, by controlling an appropriate concentration of citrate/resveratrol and gold chloride as well as stirring time and temperature. The resveratrol nanoemulsion, composed of soybean oil, Tween 80, and sucrose fatty acid ester in glycerol and water, possessed a high storage stability with a mean particle size of 14.1 nm, zeta-potential of −49.7 mV, and encapsulation efficiency of 95.5%. An antiproliferation study revealed that both R-GNPs and resveratrol nanoemulsion could effectively inhibit the growth of pancreatic cancer cells BxPC-3, with the latter showing a higher inhibition effect. Western blot analysis implied that both can down-regulate expressions of cyclin A, cyclin B, CDK1, and CDK2 and up-regulate expressions of p53 and p21, accompanied by enhancing cytochrome C expression, decreasing BcL-2 expression, increasing Bax expression, and leading to the elevation of caspase-8, caspase-9, and caspase-3 activities for cell apoptosis execution. Future research is needed to study the inhibition of pancreatic tumors in vivo by R-GNPs and resveratrol nanoemulsions.
Collapse
Affiliation(s)
- Baskaran Stephen Inbaraj
- Department of Food Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (B.S.I.); (L.-H.H.)
| | - Leng-Huei Hua
- Department of Food Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (B.S.I.); (L.-H.H.)
| | - Bing-Huei Chen
- Department of Food Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan; (B.S.I.); (L.-H.H.)
- Department of Nutrition, China Medical University, Taichung 40401, Taiwan
- Correspondence: ; Tel.: +886-2-2905-3626; Fax: +886-2-2209-3271
| |
Collapse
|
13
|
Zhang P, Li X, Xu Q, Wang Y, Ji J. Polydopamine nanoparticles with different sizes for NIR-promoted gene delivery and synergistic photothermal therapy. Colloids Surf B Biointerfaces 2021; 208:112125. [PMID: 34601352 DOI: 10.1016/j.colsurfb.2021.112125] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022]
Abstract
The combination of photothermal therapy and gene therapy has received increasing attention in tumor treatment. However, how to improve synergistic efficacy has become a new challenge. NIR light has a great potential in tumor treatment because of its considerable penetration depth and spatiotemporal controllability. Polydopamine is a popular photothermal conversion agent, which has desirable photothermal conversion ability and good biocompatibility. In this research, polydopamine-polyethyleneimine nanoparticles with diameters of 13 nm (SPPNPs) and 236 nm (LPPNPs) were prepared as gene carriers. The size of polydopamine nanoparticles had great effect on the complexes formation, photothermal conversion ability and gene transfection efficiency. After loading gene, the SPPNPs/gene and LPPNPs/gene complexes were about 60-80 nm and 240 nm respectively, indicating different styles of complexes formation. Both SPPNPs/gene and LPPNPs/gene complexes without NIR irradiation could achieve similar gene transfection efficiency as commercial lipofectamine 2000, while with lower cytotoxicity. Due to better photothermal conversion ability, the transfection level of LPPNPs/pGL-3 complexes increased to 4.5 times after NIR irradiation (2.6 W/cm2, 15 min), which ascribed to the quick escape of gene complexes from the endosome. The produced heat under NIR irradiation could also ablate tumor cells. So LPPNPs were chosen to deliver tumor suppressor gene p53 DNA to investigate the synergistic efficacy of gene/photothermal therapy. The tumor in KB tumor-bearing mice was almost eliminated after intratumoral injection, and the tumor inhibition efficacy of gene/photothermal synergistic therapy achieved to 99%. By combining NIR-promoted gene transfection and gene/photothermal synergistic therapy, the LPPNPs hold great promise in practical tumor treatment.
Collapse
Affiliation(s)
- Peng Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Xinfang Li
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Qinan Xu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Youxiang Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| |
Collapse
|
14
|
Chen T, Li T, Wang J. Nanoscale Au@SiO 2-drug/VEGF as an in vivo probe for osteosarcoma diagnosis and therapy. Oncol Lett 2021; 22:766. [PMID: 34589145 PMCID: PMC8442140 DOI: 10.3892/ol.2021.13027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/05/2021] [Indexed: 11/05/2022] Open
Abstract
Osteosarcoma is a common primary bone malignancy, with a 5-year survival rate of only 20–30% in patients undergoing surgical treatment. Thus, it is important to identify novel methods for diagnosing and treating osteosarcoma, which was the aim of the present study. Vascular endothelial growth factor (VEGF) was used as the tumor-targeting protein to synthesize a multifunctional core-shell nanostructure, Au@SiO2-drug/VEGF, in which the drug can be indocyanine green (ICG; as an optical tracer) or doxorubicin (DOX; as a chemotherapeutic agent). With VEGF as the osteosarcoma-targeting protein, Au exhibited optimal photothermal transformation performance, while SiO2 served as the carrier for the drug. Au@SiO2-ICG/VEGF nanoparticles (NPs) were evaluated for imaging and for the monitoring of drug accumulation in a tumor region in mice. Once the optimal drug accumulation was achieved, combined treatment of osteosarcoma (chemotherapy and photothermal therapy) was assessed. In the perioperative period associated with minimal invasive embolization of osteosarcoma, photothermal therapy and chemotherapy were applied for osteosarcoma diagnosis using Au@SiO2-DOX/VEGF NPs. Taken together, the results of the present study provide a promising strategy for tumor detection prior to surgical treatment to improve the survival outcome of patients with osteosarcoma.
Collapse
Affiliation(s)
- Tiangui Chen
- Department of Orthopedics Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Tianbo Li
- Department of Orthopedics Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| | - Jiangning Wang
- Department of Orthopedics Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, P.R. China
| |
Collapse
|
15
|
Rinoldi C, Zargarian SS, Nakielski P, Li X, Liguori A, Petronella F, Presutti D, Wang Q, Costantini M, De Sio L, Gualandi C, Ding B, Pierini F. Nanotechnology-Assisted RNA Delivery: From Nucleic Acid Therapeutics to COVID-19 Vaccines. SMALL METHODS 2021; 5:e2100402. [PMID: 34514087 PMCID: PMC8420172 DOI: 10.1002/smtd.202100402] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/04/2021] [Indexed: 05/07/2023]
Abstract
In recent years, the main quest of science has been the pioneering of the groundbreaking biomedical strategies needed for achieving a personalized medicine. Ribonucleic acids (RNAs) are outstanding bioactive macromolecules identified as pivotal actors in regulating a wide range of biochemical pathways. The ability to intimately control the cell fate and tissue activities makes RNA-based drugs the most fascinating family of bioactive agents. However, achieving a widespread application of RNA therapeutics in humans is still a challenging feat, due to both the instability of naked RNA and the presence of biological barriers aimed at hindering the entrance of RNA into cells. Recently, material scientists' enormous efforts have led to the development of various classes of nanostructured carriers customized to overcome these limitations. This work systematically reviews the current advances in developing the next generation of drugs based on nanotechnology-assisted RNA delivery. The features of the most used RNA molecules are presented, together with the development strategies and properties of nanostructured vehicles. Also provided is an in-depth overview of various therapeutic applications of the presented systems, including coronavirus disease vaccines and the newest trends in the field. Lastly, emerging challenges and future perspectives for nanotechnology-mediated RNA therapies are discussed.
Collapse
Affiliation(s)
- Chiara Rinoldi
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Seyed Shahrooz Zargarian
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Pawel Nakielski
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| | - Xiaoran Li
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Anna Liguori
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
| | - Francesca Petronella
- Institute of Crystallography CNR‐ICNational Research Council of ItalyVia Salaria Km 29.300Monterotondo – Rome00015Italy
| | - Dario Presutti
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Qiusheng Wang
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Marco Costantini
- Institute of Physical ChemistryPolish Academy of Sciencesul. M. Kasprzaka 44/52Warsaw01‐224Poland
| | - Luciano De Sio
- Department of Medico‐Surgical Sciences and BiotechnologiesResearch Center for BiophotonicsSapienza University of RomeCorso della Repubblica 79Latina04100Italy
- CNR‐Lab. LicrylInstitute NANOTECArcavacata di Rende87036Italy
| | - Chiara Gualandi
- Department of Chemistry “Giacomo Ciamician” and INSTM UdR of BolognaUniversity of BolognaVia Selmi 2Bologna40126Italy
- Interdepartmental Center for Industrial Research on Advanced Applications in Mechanical Engineering and Materials TechnologyCIRI‐MAMUniversity of BolognaViale Risorgimento 2Bologna40136Italy
| | - Bin Ding
- Innovation Center for Textile Science and TechnologyDonghua UniversityWest Yan'an Road 1882Shanghai200051China
| | - Filippo Pierini
- Department of Biosystems and Soft MatterInstitute of Fundamental Technological ResearchPolish Academy of Sciencesul. Pawińskiego 5BWarsaw02‐106Poland
| |
Collapse
|
16
|
Wang P, Zhou Y, Richards AM. Effective tools for RNA-derived therapeutics: siRNA interference or miRNA mimicry. Theranostics 2021; 11:8771-8796. [PMID: 34522211 PMCID: PMC8419061 DOI: 10.7150/thno.62642] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
The approval of the first small interfering RNA (siRNA) drug Patisiran by FDA in 2018 marks a new era of RNA interference (RNAi) therapeutics. MicroRNAs (miRNA), an important post-transcriptional gene regulator, are also the subject of both basic research and clinical trials. Both siRNA and miRNA mimics are ~21 nucleotides RNA duplexes inducing mRNA silencing. Given the well performance of siRNA, researchers ask whether miRNA mimics are unnecessary or developed siRNA technology can pave the way for the emergence of miRNA mimic drugs. Through comprehensive comparison of siRNA and miRNA, we focus on (1) the common features and lessons learnt from the success of siRNAs; (2) the unique characteristics of miRNA that potentially offer additional therapeutic advantages and opportunities; (3) key areas of ongoing research that will contribute to clinical application of miRNA mimics. In conclusion, miRNA mimics have unique properties and advantages which cannot be fully matched by siRNA in clinical applications. MiRNAs are endogenous molecules and the gene silencing effects of miRNA mimics can be regulated or buffered to ameliorate or eliminate off-target effects. An in-depth understanding of the differences between siRNA and miRNA mimics will facilitate the development of miRNA mimic drugs.
Collapse
Affiliation(s)
- Peipei Wang
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Medicine, National University Health System, 119228 Singapore
| | - Yue Zhou
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Medicine, National University Health System, 119228 Singapore
| | - Arthur M. Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Medicine, National University Health System, 119228 Singapore
- Christchurch Heart Institute, Department of Medicine, University of Otago Christchurch, New Zealand
| |
Collapse
|
17
|
Wang L, Yan Y. A Review of pH-Responsive Organic-Inorganic Hybrid Nanoparticles for RNAi-Based Therapeutics. Macromol Biosci 2021; 21:e2100183. [PMID: 34160896 DOI: 10.1002/mabi.202100183] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/04/2021] [Indexed: 12/13/2022]
Abstract
RNA interference (RNAi) shows great potential in the treatment of varying cancer and genetic disorders. The lack of safe and effective delivery methods is an ongoing challenge to realize the full potential of RNAi-based therapeutics. pH-responsive hybrid nanoparticle is a promising non-virus platform for small interfering RNA (siRNA) delivery with unique properties including the robust response to the acidic microenvironment and the capability of theranostic and combined therapeutics. The mechanism of RNAi and the delivery barriers for RNAi-based therapeutics are first discussed. Then, the general patterns of pH-response and the typical construction of hybrid nanoparticles are demonstrated. The recent advances in pH-responsive organic-inorganic hybrid nanoparticles for siRNA delivery are highlighted, in particular, how pH-response of ionizable groups, acid-labile bonds, and decomposition of inorganic components affect the physicochemical properties of hybrid nanoparticles and benefit the cellular uptake and intracellular trafficking of siRNA payloads are discussed. At last, the remaining problems and the prospects for pH-responsive hybrid nanoparticles for siRNA delivery are analyzed.
Collapse
Affiliation(s)
- Lu Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yunfeng Yan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
18
|
Promise of gold nanomaterials as a lung cancer theranostic agent: a systematic review. INTERNATIONAL NANO LETTERS 2021. [DOI: 10.1007/s40089-021-00332-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
19
|
Torres-Vanegas JD, Cruz JC, Reyes LH. Delivery Systems for Nucleic Acids and Proteins: Barriers, Cell Capture Pathways and Nanocarriers. Pharmaceutics 2021; 13:428. [PMID: 33809969 PMCID: PMC8004853 DOI: 10.3390/pharmaceutics13030428] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 12/27/2022] Open
Abstract
Gene therapy has been used as a potential approach to address the diagnosis and treatment of genetic diseases and inherited disorders. In this line, non-viral systems have been exploited as promising alternatives for delivering therapeutic transgenes and proteins. In this review, we explored how biological barriers are effectively overcome by non-viral systems, usually nanoparticles, to reach an efficient delivery of cargoes. Furthermore, this review contributes to the understanding of several mechanisms of cellular internalization taken by nanoparticles. Because a critical factor for nanoparticles to do this relies on the ability to escape endosomes, researchers have dedicated much effort to address this issue using different nanocarriers. Here, we present an overview of the diversity of nanovehicles explored to reach an efficient and effective delivery of both nucleic acids and proteins. Finally, we introduced recent advances in the development of successful strategies to deliver cargoes.
Collapse
Affiliation(s)
- Julian D. Torres-Vanegas
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos (GDPP), Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| |
Collapse
|
20
|
Zou M, Du Y, Liu R, Zheng Z, Xu J. Nanocarrier-delivered small interfering RNA for chemoresistant ovarian cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1648. [PMID: 33682310 DOI: 10.1002/wrna.1648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States. Because success in early screening is limited, and most patients with advanced disease develop resistance to multiple treatment modalities, the overall prognosis of ovarian cancer is poor. Despite the revolutionary role of surgery and chemotherapy in curing ovarian cancer, recurrence remains a major challenge in treatment. Thus, improving our understanding of the pathogenesis of ovarian cancer is essential for developing more effective treatments. In this review, we analyze the underlying molecular mechanisms leading to chemotherapy resistance. We discuss the clinical benefits and potential challenges of using nanocarrier-delivered small interfering RNA to treat chemotherapy-resistant ovarian cancer. We aim to elicit collaborative studies on nanocarrier-delivered small interfering RNA to improve the long-term survival rate and quality of life of patients with ovarian cancer. This article is categorized under: RNA Methods > RNA Nanotechnology Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Mingyuan Zou
- Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruizhen Liu
- The First People's Hospital of Wu'an, Wu'an, Hebei, China
| | - Zeliang Zheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Izci M, Maksoudian C, Manshian BB, Soenen SJ. The Use of Alternative Strategies for Enhanced Nanoparticle Delivery to Solid Tumors. Chem Rev 2021; 121:1746-1803. [PMID: 33445874 PMCID: PMC7883342 DOI: 10.1021/acs.chemrev.0c00779] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Indexed: 02/08/2023]
Abstract
Nanomaterial (NM) delivery to solid tumors has been the focus of intense research for over a decade. Classically, scientists have tried to improve NM delivery by employing passive or active targeting strategies, making use of the so-called enhanced permeability and retention (EPR) effect. This phenomenon is made possible due to the leaky tumor vasculature through which NMs can leave the bloodstream, traverse through the gaps in the endothelial lining of the vessels, and enter the tumor. Recent studies have shown that despite many efforts to employ the EPR effect, this process remains very poor. Furthermore, the role of the EPR effect has been called into question, where it has been suggested that NMs enter the tumor via active mechanisms and not through the endothelial gaps. In this review, we provide a short overview of the EPR and mechanisms to enhance it, after which we focus on alternative delivery strategies that do not solely rely on EPR in itself but can offer interesting pharmacological, physical, and biological solutions for enhanced delivery. We discuss the strengths and shortcomings of these different strategies and suggest combinatorial approaches as the ideal path forward.
Collapse
Affiliation(s)
- Mukaddes Izci
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Christy Maksoudian
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational
Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth
and Optical Imaging Group, Translational Cell and Tissue Research
Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| |
Collapse
|
22
|
Dang MN, Gomez Casas C, Day ES. Photoresponsive miR-34a/Nanoshell Conjugates Enable Light-Triggered Gene Regulation to Impair the Function of Triple-Negative Breast Cancer Cells. NANO LETTERS 2021; 21:68-76. [PMID: 33306406 PMCID: PMC7855941 DOI: 10.1021/acs.nanolett.0c03152] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive disease that requires new interventions. A promising approach to improve patient prognosis is to introduce tumor suppressive miR-34a into TNBC cells. Unfortunately, naked miR-34a is not effective therapeutically because it is degraded by nucleases and cannot passively enter cells. Nanocarriers designed to increase miR-34a stability and cellular entry have lacked specificity and potency. To overcome these limitations, we conjugated miR-34a to photoresponsive gold nanoshells (NS), which can release tethered miR-34a upon excitation with continuous wave (CW) or nanosecond (ns) pulsed near-infrared light to facilitate on-demand gene regulation. We demonstrate that miR-34a/NS can regulate downstream miR-34a targets following irradiation to reduce TNBC cell viability, proliferation, and migration. Further, we show ns pulsed light releases miRNA more effectively than CW light, and that released miR-34a is as potent as transfected miR-34a. These findings signify miR-34a/NS as promising tools for precisely controlled gene regulation of TNBC.
Collapse
Affiliation(s)
- Megan N Dang
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Carolina Gomez Casas
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Emily S Day
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Helen F. Graham Cancer Center & Research Institute, Newark, Delaware 19713, United States
| |
Collapse
|
23
|
Yasun E, Gandhi S, Choudhury S, Mohammadinejad R, Benyettou F, Gozubenli N, Arami H. Hollow micro and nanostructures for therapeutic and imaging applications. J Drug Deliv Sci Technol 2020; 60:102094. [PMID: 34335877 PMCID: PMC8320649 DOI: 10.1016/j.jddst.2020.102094] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hollow particles have been extensively used in bioanalytical and biomedical applications for almost two decades due to their unique and tunable optoelectronic properties as well as their significantly high loading capacities. These intrinsic properties led them to be used in various bioimaging applications as contrast agents, controlled delivery (i.e. drugs, nucleic acids and other biomolecules) platforms and photon-triggered therapies (e.g. photothermal and photodynamic therapies). Since recent studies showed that imaging-guided targeted therapeutics have higher success rates, multimodal theranostic platforms (combination of one or more therapy and diagnosis modality) have been employed more often and hollow particles (i.e. nanoshells) have been one of the most efficient candidates to be used in multiple-purpose platforms, owing to their intrinsic properties that enable synergistic multimodal performance. In this review, recent advances in the applications of such hollow particles fabricated with various routes (either inorganic or organic based) were summarized to delineate strategies for tuning their properties for more efficient biomedical performance by overcoming common biological barriers. This review will pave the ways for expedited progress in design of next generation of hollow particles for clinical applications.
Collapse
Affiliation(s)
- Emir Yasun
- University of California, Santa Barbara and California NanoSystems Institute (CNSI), Santa Barbara, CA, 93106, USA
| | - Sonu Gandhi
- DBT-National Institute of Animal Biotechnology, Hyderabad, 500032, Telangana, India
| | - Samraggi Choudhury
- DBT-National Institute of Animal Biotechnology, Hyderabad, 500032, Telangana, India
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Farah Benyettou
- New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Numan Gozubenli
- Molecular Biology and Genetics Department, Harran University, Sanliurfa, Turkey
| | - Hamed Arami
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
- Molecular Imaging Program at Stanford (MIPS), The James H Clark Center, Stanford University, Stanford, CA, USA
| |
Collapse
|
24
|
Wen H, Tamarov K, Happonen E, Lehto V, Xu W. Inorganic Nanomaterials for Photothermal‐Based Cancer Theranostics. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000207] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Huang Wen
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Konstantin Tamarov
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Emilia Happonen
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Vesa‐Pekka Lehto
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| | - Wujun Xu
- Department of Applied Physics University of Eastern Finland Kuopio 70211 Finland
| |
Collapse
|
25
|
Choi SK. Photoactivation Strategies for Therapeutic Release in Nanodelivery Systems. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000117] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Seok Ki Choi
- Michigan Nanotechnology Institute for Medicine and Biological Sciences University of Michigan Medical School Ann Arbor MI 48109 USA
- Department of Internal Medicine University of Michigan Medical School Ann Arbor MI 48109 USA
| |
Collapse
|
26
|
Zhang F, Wu Q, Liu H. NIR light-triggered nanomaterials-based prodrug activation towards cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1643. [PMID: 32394638 DOI: 10.1002/wnan.1643] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 01/10/2023]
Abstract
Nanomaterials-based prodrug activation systems have been widely explored in cancer therapy, aiming at overcoming limited dosage formulation, systemic toxicity, and insufficient pharmacokinetic performance of parent drugs. For better delivery control, various stimuli systems, especially nanomaterials-based ones, have come to the forefront. Among them, near-infrared (NIR) light takes advantage of on-demand/site-specific regulation and non-invasiveness. In this review, we will address the developments of nanomaterials-based prodrug over the last decade, the activation mechanisms, and bioapplications under NIR light triggering. The advantages and limitations of NIR-triggered prodrug activation strategies and the perspectives of the next-generation prodrug nanomedicine will also be summarized. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Fengrong Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, China
| | - Qingyuan Wu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, China
| | - Huiyu Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Bionanomaterials & Translational Engineering Laboratory, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
27
|
Gold Nanoparticles in Glioma Theranostics. Pharmacol Res 2020; 156:104753. [PMID: 32209363 DOI: 10.1016/j.phrs.2020.104753] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 01/07/2023]
Abstract
Despite many endeavors to treat malignant gliomas in the last decades, the median survival of patients has not significantly improved. The infiltrative nature of high-grade gliomas and the impermeability of the blood-brain barrier to the most therapeutic agents remain major hurdles, impeding an efficacious treatment. Theranostic platforms bridging diagnosis and therapeutic modalities aim to surmount the current limitations in diagnosis and therapy of glioma. Gold nanoparticles (AuNPs) due to their biocompatibility and tunable optical properties have widely been utilized for an assortment of theranostic purposes. In this Review, applications of AuNPs as imaging probes, drug/gene delivery systems, radiosensitizers, photothermal transducers, and multimodal theranostic agents in malignant gliomas are discussed. This Review also aims to provide a perspective on cancer theranostic applications of AuNPs in future clinical trials.
Collapse
|
28
|
De Matteis V, Cascione M, Toma CC, Rinaldi R. Engineered Gold Nanoshells Killing Tumor Cells: New Perspectives. Curr Pharm Des 2020; 25:1477-1489. [PMID: 31258061 DOI: 10.2174/1381612825666190618155127] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/11/2019] [Indexed: 12/30/2022]
Abstract
The current strategies to treat different kinds of cancer are mainly based on chemotherapy, surgery and radiation therapy. Unfortunately, these approaches are not specific and rather invasive as well. In this scenario, metal nano-shells, in particular gold-based nanoshells, offer interesting perspectives in the effort to counteract tumor cells, due to their unique ability to tune Surface Plasmon Resonance in different light-absorbing ranges. In particular, the Visible and Near Infrared Regions of the electromagnetic spectrum are able to penetrate through tissues. In this way, the light absorbed by the gold nanoshell at a specific wavelength is converted into heat, inducing photothermal ablation in treated cancer cells. Furthermore, inert gold shells can be easily functionalized with different types of molecules in order to bind cellular targets in a selective manner. This review summarizes the current state-of-art of nanosystems embodying gold shells, regarding methods of synthesis, bio-conjugations, bio-distribution, imaging and photothermal effects (in vitro and in vivo), providing new insights for the development of multifunctional antitumor drugs.
Collapse
Affiliation(s)
- Valeria De Matteis
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| | - Mariafrancesca Cascione
- Dipartimento di Scienze Biomediche e Oncologia Umana, Universita degli Studi di Bari "Aldo Moro", p.zza G. Cesare, c/o Policlinico, 70124 Bari, Italy
| | - Chiara C Toma
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| | - Rosaria Rinaldi
- Dipartimento di Matematica e Fisica "E. De Giorgi", Universita del Salento, Via Monteroni, 73100 Lecce, Italy
| |
Collapse
|
29
|
Sivakumar PM, Islami M, Zarrabi A, Khosravi A, Peimanfard S. Polymer-Graphene Nanoassemblies and their Applications in Cancer Theranostics. Anticancer Agents Med Chem 2019; 20:1340-1351. [PMID: 31746307 DOI: 10.2174/1871520619666191028112258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/30/2019] [Accepted: 06/03/2019] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND OBJECTIVE Graphene-based nanomaterials have received increasing attention due to their unique physical-chemical properties including two-dimensional planar structure, large surface area, chemical and mechanical stability, superconductivity and good biocompatibility. On the other hand, graphene-based nanomaterials have been explored as theranostics agents, the combination of therapeutics and diagnostics. In recent years, grafting hydrophilic polymer moieties have been introduced as an efficient approach to improve the properties of graphene-based nanomaterials and obtain new nanoassemblies for cancer therapy. METHODS AND RESULTS This review would illustrate biodistribution, cellular uptake and toxicity of polymergraphene nanoassemblies and summarize part of successes achieved in cancer treatment using such nanoassemblies. CONCLUSION The observations showed successful targeting functionality of the polymer-GO conjugations and demonstrated a reduction of the side effects of anti-cancer drugs for normal tissues.
Collapse
Affiliation(s)
- Ponnurengam M Sivakumar
- Center for Molecular Biology, Institute of Research and Development, Duy Tan University, 03 Quang Trung, Da Nang, Vietnam
| | - Matin Islami
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Orta Mah., 34956 Tuzla, Istanbul, Turkey
| | - Arezoo Khosravi
- Department of Mechanical Engineering, Khomeinishahr Branch, Islamic Azad University, Khomeinishahr/Isfahan, Iran
| | - Shohreh Peimanfard
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Isfahan, Iran
| |
Collapse
|
30
|
Ghiassian S, Yu L, Gobbo P, Nazemi A, Romagnoli T, Luo W, Luyt LG, Workentin MS. Nitrone-Modified Gold Nanoparticles: Synthesis, Characterization, and Their Potential as 18F-Labeled Positron Emission Tomography Probes via I-SPANC. ACS OMEGA 2019; 4:19106-19115. [PMID: 31763533 PMCID: PMC6868604 DOI: 10.1021/acsomega.9b02322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/28/2019] [Indexed: 05/11/2023]
Abstract
A novel bioorthogonal gold nanoparticle (AuNP) template displaying interfacial nitrone functional groups for bioorthogonal interfacial strain-promoted alkyne-nitrone cycloaddition reactions has been synthesized. These nitrone-AuNPs were characterized in detail using 1H nuclear magnetic resonance spectroscopy, transmission electron microscopy, thermogravimetric analysis, and X-ray photoelectron spectroscopy, and a nanoparticle raw formula was calculated. The ability to control the conjugation of molecules of interest at the molecular level onto the nitrone-AuNP template allowed us to create a novel methodology for the synthesis of AuNP-based radiolabeled probes.
Collapse
Affiliation(s)
- Sara Ghiassian
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Lihai Yu
- London
Regional Cancer Program, 800 Commissioners Rd. E., London N6A 5W9, Ontario, Canada
| | - Pierangelo Gobbo
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Ali Nazemi
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Tommaso Romagnoli
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Wilson Luo
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| | - Leonard G. Luyt
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
- London
Regional Cancer Program, 800 Commissioners Rd. E., London N6A 5W9, Ontario, Canada
| | - Mark S. Workentin
- Department
of Chemistry and the Center for Materials and Biomaterials
Research and Department of Oncology, The University
of Western Ontario, London N6A 5B7, Ontario, Canada
| |
Collapse
|
31
|
Nunes T, Pons T, Hou X, Van Do K, Caron B, Rigal M, Di Benedetto M, Palpant B, Leboeuf C, Janin A, Bousquet G. Pulsed-laser irradiation of multifunctional gold nanoshells to overcome trastuzumab resistance in HER2-overexpressing breast cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:306. [PMID: 31299997 PMCID: PMC6626398 DOI: 10.1186/s13046-019-1305-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 06/30/2019] [Indexed: 01/09/2023]
Abstract
Background HER2-overexpressing metastatic breast cancers are challenging practice in oncology when they become resistant to anti-HER2 therapies such as trastuzumab. In these clinical situations, HER2-overexpression persists in metastatic localizations, and can thus be used for active targeting using innovative therapeutic approaches. Functionalized gold nanoparticles with anti-HER2 antibody can be stimulated by near-infrared light to induce hyperthermia. Methods Here, hybrid anti-HER2 gold nanoshells were engineered for photothermal therapy to overcome trastuzumab resistance in HER2-overexpressing breast cancer xenografts. Results When gold nanoshells were administered in HER2-tumor xenografts, no toxicity was observed. A detailed pharmacokinetic study showed a time-dependent accumulation of gold nanoshells within the tumors, significantly greater with functionalized gold nanoshells at 72 h. This enabled us to optimize the treatment protocol and irradiate the mice when the anti-HER2 gold nanoshells had accumulated most in the tumors. After weekly injections of anti-HER2 gold nanoshells, and repeated irradiations with a femtosecond-pulsed laser over four weeks, tumor growth was significantly inhibited. Detailed tissue microscopic analyses showed that the tumor growth inhibition was due to an anti-angiogenic effect, coherent with a preferential distribution of the nanoshells in tumor microvessels. We also showed a direct tumor cell effect with apoptosis and inhibition of proliferation, coherent with an immune-mediated targeting of tumor cells by anti-HER2 nanoshells. Conclusion This preclinical study thus supports the use of anti-HER2 gold nanoshells and photothermal therapy to overcome trastuzumab resistance in HER2-overexpressing breast cancer. Electronic supplementary material The online version of this article (10.1186/s13046-019-1305-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Toni Nunes
- Université-Paris-Diderot, Sorbonne-Paris-Cité, Laboratoire Pathologie, UMR-S942, F-75010, Paris, France.,INSERM, U942, Paris, France
| | - Thomas Pons
- LPEM, ESPCI Paris, PSL Research University, CNRS, Sorbonne Universités, UPMC, F-75005, Paris, France
| | - Xue Hou
- Laboratoire de Photonique Quantique et Moléculaire, CentraleSupélec, Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, CNRS UMR 8537, 3 rue Joliot Curie, F-91190, Gif-sur-Yvette, France
| | - Khanh Van Do
- Laboratoire de Photonique Quantique et Moléculaire, CentraleSupélec, Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, CNRS UMR 8537, 3 rue Joliot Curie, F-91190, Gif-sur-Yvette, France
| | - Benoît Caron
- ALIPP6, Institut des Sciences de la Terre de Paris UMR 7193, CNRS, Sorbonne Université, F-75005, Paris, France
| | - Marthe Rigal
- AP-HP-Hôpital Avicenne, Service-Pharmacie-Paris, Paris, France
| | | | - Bruno Palpant
- Laboratoire de Photonique Quantique et Moléculaire, CentraleSupélec, Ecole Normale Supérieure Paris-Saclay, Université Paris Saclay, CNRS UMR 8537, 3 rue Joliot Curie, F-91190, Gif-sur-Yvette, France
| | - Christophe Leboeuf
- Université-Paris-Diderot, Sorbonne-Paris-Cité, Laboratoire Pathologie, UMR-S942, F-75010, Paris, France.,INSERM, U942, Paris, France
| | - Anne Janin
- Université-Paris-Diderot, Sorbonne-Paris-Cité, Laboratoire Pathologie, UMR-S942, F-75010, Paris, France. .,INSERM, U942, Paris, France. .,AP-HP-Hôpital Saint-Louis, Laboratoire-Pathologie-Paris, Paris, France.
| | - Guilhem Bousquet
- Université-Paris-Diderot, Sorbonne-Paris-Cité, Laboratoire Pathologie, UMR-S942, F-75010, Paris, France. .,INSERM, U942, Paris, France. .,Université Paris 13, F-93430, Villetaneuse, France. .,AP-HP-Hôpital Avicenne, Service-Oncologie-Paris, Paris, France.
| |
Collapse
|
32
|
Sánchez-López E, Guerra M, Dias-Ferreira J, Lopez-Machado A, Ettcheto M, Cano A, Espina M, Camins A, Garcia ML, Souto EB. Current Applications of Nanoemulsions in Cancer Therapeutics. NANOMATERIALS 2019; 9:nano9060821. [PMID: 31159219 PMCID: PMC6632105 DOI: 10.3390/nano9060821] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 05/24/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022]
Abstract
Nanoemulsions are pharmaceutical formulations composed of particles within a nanometer range. They possess the capacity to encapsulate drugs that are poorly water soluble due to their hydrophobic core nature. Additionally, they are also composed of safe gradient excipients, which makes them a stable and safe option to deliver drugs. Cancer therapy has been an issue for several decades. Drugs developed to treat this disease are not always successful or end up failing, mainly due to low solubility, multidrug resistance (MDR), and unspecific toxicity. Nanoemulsions might be the solution to achieve efficient and safe tumor treatment. These formulations not only solve water-solubility problems but also provide specific targeting to cancer cells and might even be designed to overcome MDR. Nanoemulsions can be modified using ligands of different natures to target components present in tumor cells surface or to escape MDR mechanisms. Multifunctional nanoemulsions are being studied by a wide variety of researchers in different research areas mainly for the treatment of different types of cancer. All of these studies demonstrate that nanoemulsions are efficiently taken by the tumoral cells, reduce tumor growth, eliminate toxicity to healthy cells, and decrease migration of cancer cells to other organs.
Collapse
Affiliation(s)
- Elena Sánchez-López
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), University of Barcelona, 08028 Barcelona, Spain.
| | - Mariana Guerra
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra (FFUC), Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| | - João Dias-Ferreira
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra (FFUC), Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| | - Ana Lopez-Machado
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), University of Barcelona, 08028 Barcelona, Spain.
| | - Miren Ettcheto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra (FFUC), Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Amanda Cano
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), University of Barcelona, 08028 Barcelona, Spain.
| | - Marta Espina
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain.
| | - Antoni Camins
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra (FFUC), Polo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain.
| | - Maria Luisa Garcia
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), University of Barcelona, 08028 Barcelona, Spain.
| | - Eliana B Souto
- Department of Pharmacy and Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy, University of Barcelona, 08028 Barcelona, Spain.
- CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
33
|
Narmani A, Rezvani M, Farhood B, Darkhor P, Mohammadnejad J, Amini B, Refahi S, Abdi Goushbolagh N. Folic acid functionalized nanoparticles as pharmaceutical carriers in drug delivery systems. Drug Dev Res 2019; 80:404-424. [PMID: 31140629 DOI: 10.1002/ddr.21545] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/02/2019] [Accepted: 05/07/2019] [Indexed: 12/15/2022]
Abstract
Conventional chemotherapeutic approaches in cancer therapy such as surgery, chemotherapy, and radiotherapy have several disadvantages due to their nontargeted distributions in the whole body. On the other hand, nanoparticles (NPs) based therapies are remarkably progressing to solve several limitations of conventional drug delivery systems (DDSs) including nonspecific biodistribution and targeting, poor water solubility, weak bioavailability and biodegradability, low pharmacokinetic properties, and so forth. The enhanced permeability and retention effect escape from P-glycoprotein trap in cancer cells as a passive targeting mechanism, and active targeting strategies are also other most important advantages of NPs in cancer diagnosis and therapy. Folic acid (FA) is one of the biologic molecules which has been targeted overexpressed-folic acid receptor (FR) on the surface of cancer cells. Therefore, conjugation of FA to NPs most easily enhances the FR-mediated targeting delivery of therapeutic agents. Here, the recent works in FA which have been decorated NPs-based DDSs are discussed and cancer therapy potency of these NPs in clinical trials are presented.
Collapse
Affiliation(s)
- Asghar Narmani
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Melina Rezvani
- Department of Biology, Faculty of Sciences, Payame Noor University, Tehran, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Parvaneh Darkhor
- Department of Medical Physics, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Mohammadnejad
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Bahram Amini
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Soheila Refahi
- Department of Medical Physics, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nouraddin Abdi Goushbolagh
- Department of Medical Physics, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
34
|
|
35
|
Alhajj N, Chee CF, Wong TW, Rahman NA, Abu Kasim NH, Colombo P. Lung cancer: active therapeutic targeting and inhalational nanoproduct design. Expert Opin Drug Deliv 2018; 15:1223-1247. [DOI: 10.1080/17425247.2018.1547280] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Nasser Alhajj
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Chin Fei Chee
- Nanotechnology & Catalysis Research Centre, University of Malaya, Kuala Lumpur, Malaysia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, iPROMISE, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Noorsaadah Abd Rahman
- Department of Chemistry, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- Wellness Research Cluster, Institute of Research Management & Monitoring, University of Malaya, Kuala Lumpur, Malaysia
| | - Paolo Colombo
- Dipartimento di Farmacia, Università degli Studi di Parma, Parma, Italy
| |
Collapse
|
36
|
Zhang N, Zhang S, Xu C, Fu L, Liu T, Zhao Y. Retracted: Decoy Oligodeoxynucleotides, Polysaccharides, and Targeted Peptide‐Functionalized Gold Nanorods for the Combined Treatment of Rheumatoid Arthritis. Adv Healthc Mater 2018; 7:e1800982. [DOI: 10.1002/adhm.201800982] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/24/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Nan Zhang
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesHeNan Province Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Advanced Pharmaceutical TechnologyMinistry of Education of ChinaHeNan Province Zhengzhou HeNan 450001 P. R. China
| | - Shasha Zhang
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Chunyu Xu
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Lingling Fu
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Tuanbing Liu
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
| | - Yongxing Zhao
- Department of PharmaceuticsSchool of Pharmaceutical SciencesZhengzhou University Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Targeting Therapy and Diagnosis for Critical DiseasesHeNan Province Zhengzhou HeNan 450001 P. R. China
- Key Laboratory of Advanced Pharmaceutical TechnologyMinistry of Education of ChinaHeNan Province Zhengzhou HeNan 450001 P. R. China
| |
Collapse
|
37
|
Light-induced mechanisms for nanocarrier's cargo release. Colloids Surf B Biointerfaces 2018; 173:825-832. [PMID: 30551298 DOI: 10.1016/j.colsurfb.2018.10.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 01/18/2023]
Abstract
Nanomaterials have been the focus of attention in several fields, including biomedicine, electronics, or catalysis, mainly due to the novel properties of the materials at the nanoscale. In the field of diagnosis, nanomaterials have been contemplated as an opportunity to improve sensitivity and time of response, therefore, facilitating early treatment and monitoring of the disease. For therapeutic applications, new drug delivery nanosystems aiming to provide enhanced efficiency have been proposed often addressing selective or controlled delivery of therapeutic agents to particular cells to maximize treatment efficacy minimizing adverse effects. The therapeutic agents can be dissolved, adsorbed, entrapped, encapsulated or attached on the surface or inside the nanocarriers. Given the context of the different generations of nanocarriers and their wide range of applications, the present article aims to discuss the nature of external stimuli which will trigger the controlled release of different biomolecules. For each class, a brief description of the physical principle, basic concepts, as well as some examples, are reported. A final discussion focused on the real implications and needs for optimal drug delivery system is presented, altogether with some considerations and prospects in the trends that diagnostics applications could follow in the next years.
Collapse
|
38
|
Zhao W, Li A, Zhang A, Zheng Y, Liu J. Recent Advances in Functional-Polymer-Decorated Transition-Metal Nanomaterials for Bioimaging and Cancer Therapy. ChemMedChem 2018; 13:2134-2149. [PMID: 30152914 DOI: 10.1002/cmdc.201800462] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/19/2018] [Indexed: 12/19/2022]
Abstract
In this review, we focus on recent advances in the synthesis of polymer-functionalized transition-metal-based nanomaterials and follow this up by discussing their applications in bioimaging diagnosis and cancer therapy. Transition-metal-based nanomaterials show great potential in cancer therapy owing to their intensive near-IR absorption, excellent photothermal conversion efficiency, strong X-ray attenuation, and magnetic properties. Functional polymers are usually introduced by a one-step or multistep method to further endow these nanomaterials with great biocompatibility and physiological stability. Polymer-decorated transition-metal nanomaterials show great potential in multimodal imaging diagnosis (photoacoustic imaging, computed tomography, photoluminescence imaging, positron emission tomography, etc.) and cancer therapy (chemotherapy, photothermal therapy, microwave therapy, radiotherapy, photodynamic therapy). At the end of this review, the prospects of these polymer-decorated transition-metal-based nanomaterials are also discussed.
Collapse
Affiliation(s)
- Wei Zhao
- College of Materials Science and Engineering, Institutor for Graphene Applied Technology Innovation, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Aihua Li
- College of Materials Science and Engineering, Institutor for Graphene Applied Technology Innovation, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Aitang Zhang
- College of Materials Science and Engineering, Institutor for Graphene Applied Technology Innovation, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Yiwei Zheng
- College of Materials Science and Engineering, Institutor for Graphene Applied Technology Innovation, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Jingquan Liu
- College of Materials Science and Engineering, Institutor for Graphene Applied Technology Innovation, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| |
Collapse
|
39
|
Jones SK, Douglas K, Shields AF, Merkel OM. Correlating quantitative tumor accumulation and gene knockdown using SPECT/CT and bioluminescence imaging within an orthotopic ovarian cancer model. Biomaterials 2018; 178:183-192. [PMID: 29935386 PMCID: PMC6056733 DOI: 10.1016/j.biomaterials.2018.06.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/27/2018] [Accepted: 06/11/2018] [Indexed: 10/28/2022]
Abstract
Using an orthotopic model of ovarian cancer, we studied the delivery of siRNA in nanoparticles of tri-block copolymers consisting of hyperbranched polyethylenimine-graft-polycaprolactone-block-poly(ethylene glycol) (hyPEI-g-PCL-b-PEG) with and without a folic acid targeting ligand. A SKOV-3/LUC FRα overexpressing cell line was employed to mimic the clinical manifestations of ovarian cancer. Both targeted and non-targeted micelleplexes were able to effectively deliver siRNA to the primary tumor and its metastases, as measured by gamma scintillation counting and confocal microscopy. Stability of the micelleplexes was demonstrated with a serum albumin binding study. Regarding biodistribution, intravenous (I.V.) administration showed a slight advantage of FRα targeted over non-targeted micelleplex accumulation within the tumor. However, both formulations displayed significant liver uptake. On the other hand, intraperitoneally (I.P.) injected mice showed a modest 6% of the injected dose per gram (ID/g) uptake within the primary and most interestingly also in the metastatic lesions which subsequently resulted in a 62% knockdown of firefly luciferase expression in the tumor after a single injection. While this is, to the best of our knowledge, the first paper that correlates quantitative tumor accumulation in an orthotopic tumor model with in vivo gene silencing, these data demonstrate that PEI-g-PCL-b-PEG-Fol conjugates are a promising option for gene knockdown in ovarian cancer.
Collapse
Affiliation(s)
- Steven K Jones
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kirk Douglas
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Anthony F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Olivia M Merkel
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA; Department of Pharmaceutical Sciences, Wayne State University School of Pharmacy and Health Sciences, Detroit, MI, USA; Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
40
|
Wang F, Huang Q, Wang Y, Zhang W, Lin R, Yu Y, Shen Y, Cui H, Guo S. Rational design of multimodal therapeutic nanosystems for effective inhibition of tumor growth and metastasis. Acta Biomater 2018; 77:240-254. [PMID: 30012354 DOI: 10.1016/j.actbio.2018.07.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/26/2018] [Accepted: 07/12/2018] [Indexed: 12/28/2022]
Abstract
Simultaneous inhibition of both tumor growth and metastasis is the key to treating metastatic cancer, yet the development of effective drug delivery systems represents a great challenge since multimodal therapeutic agents must be rationally combined to overcome the biological mechanisms underpinning tumor cell proliferation and invasion. In this context, we report a hybrid therapeutic nanoscale platform that incorporates an anti-proliferative drug, doxorubicin (DOX), and an anti-NF-κB agent, p65-shRNA, for effective treatment of metastatic breast cancer. In our design, we first conjugated DOX via an acid-labile linker onto gold nanorods that were pre-modified with the tumor targeting peptide RGD and a positively charged, disulfide cross-linked short polyethylenimines (DSPEI), and then incorporated shRNA through electrostatic complexation with DSPEI. We show that this "all in one" nanotherapeutic system (RDG/shRNA@DOX) can be effectively internalized through RGD-mediated endocytosis, followed by stimuli-responsive intracellular co-release of DOX and shRNA. Our in vitro experiments suggest that this multimodal system can significantly inhibit cell proliferation, angiogenesis, and invasion of metastatic MDA-MB-435 cancer cells. Systemic administration of RDG/shRNA@DOX into a metastatic mouse model led to enhanced tumor accumulation, and, most importantly, significant inhibition of in situ tumor growth and almost complete suppression of tumor metastasis. We believe this hybrid multimodal nanotherapeutic system provides important insight into the rational design of therapeutic systems for the effective treatment of metastatic carcinoma. STATEMENT OF SIGNIFICANCE The key to successfully treat metastatic cancer is the simultaneous inhibition of both tumor growth and metastasis. This represents a great challenge for the design of drug delivery systems since multimodal therapeutic agents must be rationally combined to overcome the respective biological mechanisms underpinning tumor cell proliferation and invasion. Toward this end, we developed a hybrid nanomedicine platform that incorporates an anti-proliferative drug, doxorubicin (DOX), and an anti-NF-κB agent, p65-shRNA, for effective treatment of metastatic breast cancer. We showed that this multimodal system (RDG/shRNA@DOX) enhanced tumor accumulation, led to prolonged circulation, and most importantly, significant inhibition of in situ tumor growth and almost complete suppression of tumor metastasis. We believe this hybrid multimodal nanotherapeutic system provides significant insight into the rational design of therapeutic systems for the effective treatment of metastatic cancer.
Collapse
Affiliation(s)
- Feihu Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, MD 21218, United States
| | - Qian Huang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Yun Wang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Wenjun Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Ran Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, MD 21218, United States
| | - Yanna Yu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Yuanyuan Shen
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, MD 21218, United States; Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States.
| | - Shengrong Guo
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
41
|
Banstola A, Emami F, Jeong JH, Yook S. Current Applications of Gold Nanoparticles for Medical Imaging and as Treatment Agents for Managing Pancreatic Cancer. Macromol Res 2018. [DOI: 10.1007/s13233-018-6139-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
42
|
Xiong C, Lu W, Zhou M, Wen X, Li C. Cisplatin-loaded hollow gold nanoparticles for laser-triggered release. Cancer Nanotechnol 2018; 9:6. [PMID: 30147806 PMCID: PMC6096947 DOI: 10.1186/s12645-018-0041-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/18/2018] [Indexed: 01/24/2023] Open
Abstract
Background Hollow gold nanoparticles (HGNPs) exposed to near-infrared (NIR) light yield photothermal effects that can trigger a variety of biological effects for potential biomedical applications. However, the mechanism of laser-triggered drug release has not been studied before. Methods A tripeptide Ac-Glu-Glu-Cys-NH2 (Ac-EEC) was directly linked to the surface of HGNPs. The EEC-HGNPs conjugate was then complexed with cisplatin Pt(II) to give Ac-EEC(Pt)-HGNPs. Folic acid was introduced to the gold surface of Ac-EEC-HGNPs through a thioctic acid-terminated polyethylene glycol linker (F-PEG-TA) followed by complexation with Pt(II) to give F-Ac-EEC(Pt)-HGNPs. Laser treatment was instituted with a 15-ns pulsed laser at a repetition rate of 10 Hz. The released Pt(II) was quantified by inductively coupled plasma mass spectroscopy, and the nature of the released Pt-containing species was characterized by liquid chromatography-mass spectroscopy. The cytotoxicity was studied using the MTT assay. Results Pt(II) was released from Ac-EEC(Pt)-HGNPs via two modes: (1) sustained release through an inverse ligand exchange reaction with chloride ions and (2) rapid release through cleavage of the Au-S bond between the tripeptide linker and Au surface upon NIR laser irradiation. The folate (F) conjugate of the nanoconstruct, F-Ac-EEC(Pt)-HGNPs, in combination with laser treatment showed a significantly greater effect on cell mortality against folate-overexpressing human epidermoid carcinoma KB cells than F-Ac-ECC(Pt)-HGNPs alone after 24 h of incubation. Conclusions These results demonstrate that the photothermal property of HGNPs can be used for dual-modality photothermal therapy and NIR laser-triggered platinum-based chemotherapy.
Collapse
Affiliation(s)
- Chiyi Xiong
- 1Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Wei Lu
- 1Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, USA.,2Present Address: School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai, 201203 China
| | - Min Zhou
- 1Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, USA.,3Present Address: Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310029 China
| | - Xiaoxia Wen
- 1Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Chun Li
- 1Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
43
|
The Role of the Nuclear Factor κB Pathway in the Cellular Response to Low and High Linear Energy Transfer Radiation. Int J Mol Sci 2018; 19:ijms19082220. [PMID: 30061500 PMCID: PMC6121395 DOI: 10.3390/ijms19082220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/24/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Astronauts are exposed to considerable doses of space radiation during long-term space missions. As complete shielding of the highly energetic particles is impracticable, the cellular response to space-relevant radiation qualities has to be understood in order to develop countermeasures and to reduce radiation risk uncertainties. The transcription factor Nuclear Factor κB (NF-κB) plays a fundamental role in the immune response and in the pathogenesis of many diseases. We have previously shown that heavy ions with a linear energy transfer (LET) of 100–300 keV/µm have a nine times higher potential to activate NF-κB compared to low-LET X-rays. Here, chemical inhibitor studies using human embryonic kidney cells (HEK) showed that the DNA damage sensor Ataxia telangiectasia mutated (ATM) and the proteasome were essential for NF-κB activation in response to X-rays and heavy ions. NF-κB’s role in cellular radiation response was determined by stable knock-down of the NF-κB subunit RelA. Transfection of a RelA short-hairpin RNA plasmid resulted in higher sensitivity towards X-rays, but not towards heavy ions. Reverse Transcriptase real-time quantitative PCR (RT-qPCR) showed that after exposure to X-rays and heavy ions, NF-κB predominantly upregulates genes involved in intercellular communication processes. This process is strictly NF-κB dependent as the response is completely absent in RelA knock-down cells. NF-κB’s role in the cellular radiation response depends on the radiation quality.
Collapse
|
44
|
Yang X, Fan B, Gao W, Li L, Li T, Sun J, Peng X, Li X, Wang Z, Wang B, Zhang R, Xie J. Enhanced endosomal escape by photothermal activation for improved small interfering RNA delivery and antitumor effect. Int J Nanomedicine 2018; 13:4333-4344. [PMID: 30087564 PMCID: PMC6061202 DOI: 10.2147/ijn.s161908] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Effective endosomal escape is still a critical bottleneck for intracellular delivery of small interfering RNAs (siRNAs) to maximize their therapeutic efficacy. To overcome this obstacle, we have developed a photothermally triggered system by using the near-infrared (NIR) irradiation to achieve "on-demand" endosomal escape and subsequent siRNA release into cytoplasm. MATERIALS AND METHODS Herein, the poly-L-lysine (PLL) was successfully conjugated with melanin to obtain melanin-poly-L-lysine (M-PLL) polymer as a siRNA vehicle. The melanin was an efficient photothermal sensitizer, and the positive pendant amino groups of PLL could condense siRNAs to form stable complexes by electrostatic interactions. RESULTS AND DISCUSSION Inspired by its excellent photothermal conversion efficiency, the melanin was first involved in the siRNA delivery system. Confocal laser scanning microscopic observation revealed that after cellular uptake the photothermally induced endosomal escape could facilitate siRNAs to overcome endosomal barrier and be delivered into cytoplasm, which resulted in significant silence in the luciferase expression over the NIR- and melanin-free controls. Moreover, the anti-survivin siRNA-loaded M-PLL nanoparticles displayed great inhibitory effect on 4T1 tumor growth in vitro and in vivo. CONCLUSION These findings suggest that the M-PLL-mediated siRNA delivery is a promising candidate for therapeutic siRNA delivery and shows improved effect for cancer therapy via enhanced endosomal escape.
Collapse
Affiliation(s)
- Xi Yang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
- Imaging Department of Shanxi Provincial Cancer Hospital, Shanxi Medical University, Imaging Department of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| | - Bo Fan
- Department of Pharmacy, School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Wei Gao
- Department of Otolaryngology, Head and Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, People's Republic of China
| | - Liping Li
- Imaging Department of Shanxi Provincial Cancer Hospital, Shanxi Medical University, Imaging Department of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| | - Tingting Li
- Department of Pharmacy, School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
| | - Jinghua Sun
- Imaging Department of Shanxi Provincial Cancer Hospital, Shanxi Medical University, Imaging Department of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| | - Xiaoyang Peng
- Imaging Department of Shanxi Provincial Cancer Hospital, Shanxi Medical University, Imaging Department of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| | - Xiaoyan Li
- Imaging Department of Shanxi Provincial Cancer Hospital, Shanxi Medical University, Imaging Department of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| | - Zhenjun Wang
- Imaging Department of Shanxi Provincial Cancer Hospital, Shanxi Medical University, Imaging Department of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| | - Binquan Wang
- Department of Otolaryngology, Head and Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, People's Republic of China
| | - Ruiping Zhang
- Imaging Department of Shanxi Provincial Cancer Hospital, Shanxi Medical University, Imaging Department of Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, Shanxi, People's Republic of China,
| |
Collapse
|
45
|
Tan X, Burchfield EL, Zhang K. Light-responsive Drug Delivery Systems. STIMULI-RESPONSIVE DRUG DELIVERY SYSTEMS 2018. [DOI: 10.1039/9781788013536-00163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Materials that interact with light and subsequently change their physicochemical properties are of great interest for drug delivery. The human body is semitransparent to light of the near-infrared (NIR) region, which makes it possible to use light as an external stimulus to trigger drug release. In this chapter, we review light-triggered drug release systems of both photochemical and photothermal mechanisms. We explore recent literature on a variety of light-responsive materials for drug delivery, including organic, inorganic, and hybrid systems, which collectively embody the strategies for synergizing light responsiveness for controlled drug release/activation with other drug delivery techniques.
Collapse
Affiliation(s)
- X. Tan
- Northeastern University, Department of Chemistry and Chemical Biology 360 Huntington Ave. Boston MA 02115 USA
| | - E. L. Burchfield
- Northeastern University, Department of Chemistry and Chemical Biology 360 Huntington Ave. Boston MA 02115 USA
| | - K. Zhang
- Northeastern University, Department of Chemistry and Chemical Biology 360 Huntington Ave. Boston MA 02115 USA
| |
Collapse
|
46
|
Mao X, Tong J, Wang Y, Zhu Z, Yin Y, Wang Y. Triptolide exhibits antitumor effects by reversing hypermethylation of WIF‑1 in lung cancer cells. Mol Med Rep 2018; 18:3041-3049. [PMID: 30015908 DOI: 10.3892/mmr.2018.9263] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 03/21/2018] [Indexed: 11/05/2022] Open
Abstract
Triptolide (TP) exhibits numerous biological activities, including immunosuppressive, anti‑inflammatory and antitumor effects. The aim of the present study was to investigate the role of TP as a potent therapeutic drug for the treatment of lung cancer and to investigate the underlying therapeutic mechanisms. Western blot analyses and reverse transcription‑quantitative polymerase chain reaction (PCR) were performed to investigate the expression of genes at transcriptional and translational levels, respectively. Methylation‑specific PCR assays were conducted to investigate whether TP affects the Wnt inhibitory factor‑1 (WIF‑1) methylation status and subsequently affects apoptosis, migration or the invasion of lung cancer cells. The results of the present study revealed that the methylation status of WIF‑1 in lung cancer cell lines A549 and H460 was significantly enhanced compared with the human normal bronchial epithelial cell line HBE, whereas treatment with TP was revealed to induce the demethylation of WIF‑1. The present study aimed to investigate whether the biological activities of TP are regulated by inhibiting the Wnt signaling pathway via an increase in WIF‑1 expression levels. The results of the present study revealed that Wnt signaling was suppressed in cells following treatment with TP, which was concluded by the downregulation of Axin 2 and β‑catenin expression. Further investigation demonstrated that the silencing of WIF‑1 expression with small interfering RNA reversed the TP‑induced upregulation of WIF‑1 expression, upregulated Axin 2 and β‑catenin expression and enhanced the activation of Wnt signaling. Notably, an upregulation of cellular tumor antigen p53 expression, and downregulation of matrix metalloproteinase‑9 (MMP‑9) and phosphorylated‑nuclear factor‑κB (NF‑κB) P65 (p‑P65) levels was observed following TP treatment. These results suggest that the Wnt, p53 and NF‑κB signaling pathways mediate the potent antitumor effects of TP. Notably, the silencing of WIF‑1 did not completely recover the levels of p53, MMP‑9 and p‑P65 in cells treated with TP compared with the control cells, thus suggesting that TP exhibits further functions in addition to the targeting of WIF‑1.
Collapse
Affiliation(s)
- Xiaoliang Mao
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Jichun Tong
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yong Wang
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Zheng Zhu
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yajun Yin
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| | - Yeming Wang
- Department of Cardiothoracic Surgery, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
47
|
Evans ER, Bugga P, Asthana V, Drezek R. Metallic Nanoparticles for Cancer Immunotherapy. MATERIALS TODAY (KIDLINGTON, ENGLAND) 2018; 21:673-685. [PMID: 30197553 PMCID: PMC6124314 DOI: 10.1016/j.mattod.2017.11.022] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cancer immunotherapy, or the utilization of the body's immune system to attack tumor cells, has gained prominence over the past few decades as a viable cancer treatment strategy. Recently approved immunotherapeutics have conferred remission upon patients with previously bleak outcomes and have expanded the number of tools available to treat cancer. Nanoparticles -including polymeric, liposomal, and metallic formulations - naturally traffic to the spleen and lymph organs and the relevant immune cells therein, making them good candidates for delivery of immunotherapeutic agents. Metallic nanoparticle formulations in particular are advantageous because of their potential for dense surface functionalization and their capability for optical or heat based therapeutic methods. Many research groups have investigated the potential of nanoparticle-mediated delivery platforms to improve the efficacy of immunotherapies. Despite the significant preclinical successes demonstrated by many of these platforms over the last twenty years, few metallic nanoparticles have successfully entered clinical trials with none achieving FDA approval for cancer therapy. In this review, we will discuss preclinical research and clinical trials involving metallic nanoparticles (MNPs) for cancer immunotherapy applications and discuss the potential for clinical translation of MNPs.
Collapse
Affiliation(s)
- Emily Reiser Evans
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Pallavi Bugga
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Vishwaratn Asthana
- Department of Bioengineering, Rice University, Houston, TX 77005, United States
| | - Rebekah Drezek
- Department of Bioengineering, Rice University, Houston, TX 77005, United States. Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, United States
| |
Collapse
|
48
|
Riley RS, Dang MN, Billingsley MM, Abraham B, Gundlach L, Day ES. Evaluating the Mechanisms of Light-Triggered siRNA Release from Nanoshells for Temporal Control Over Gene Regulation. NANO LETTERS 2018; 18:3565-3570. [PMID: 29701993 PMCID: PMC6450696 DOI: 10.1021/acs.nanolett.8b00681] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
The ability to regulate intracellular gene expression with exogenous nucleic acids such as small interfering RNAs (siRNAs) has substantial potential to improve the study and treatment of disease. However, most transfection agents and nanoparticle-based carriers that are used for the intracellular delivery of nucleic acids cannot distinguish between diseased and healthy cells, which may cause them to yield unintended widespread gene regulation. An ideal delivery system would only silence targeted proteins in diseased tissue in response to an external stimulus. To enable spatiotemporal control over gene silencing, researchers have begun to develop nucleic acid-nanoparticle conjugates that keep their nucleic acid cargo inactive until it is released from the nanoparticle on-demand by externally applied near-infrared laser light. This strategy can overcome several limitations of other nucleic acid delivery systems, but the mechanisms by which these platforms operate remain ill understood. Here, we perform a detailed investigation of the mechanisms by which silica core/gold shell nanoshells (NSs) release conjugated siRNA upon excitation with either pulsed or continuous wave (CW) near-infrared (NIR) light, with the goal of providing insight into how these nanoconjugates can enable on-demand gene regulation. We demonstrate that siRNA release from NSs upon pulsed laser irradiation is a temperature-independent process that is substantially more efficient than siRNA release triggered by CW irradiation. Contrary to literature, which suggests that only pulsed irradiation releases siRNA duplexes, we found that both modes of irradiation release a mixture of siRNA duplexes and single-stranded oligonucleotides, but that pulsed irradiation results in a higher percentage of released duplexes. To demonstrate that the siRNA released from NSs upon pulsed irradiation remains functional, we evaluated the use of NSs coated with green fluorescent protein (GFP)-targeted siRNA (siGFP-NS) for on-demand knockdown of GFP in cells. We found that GFP-expressing cells treated with siGFP-NS and irradiated with a pulsed laser experienced a 33% decrease in GFP expression compared to cells treated with no laser. Further, we observed that light-triggered gene silencing mediated by siGFP-NS is more potent than using commercial transfection agents to deliver siRNA into cells. This work provides unprecedented insight into the mechanisms by which plasmonic NSs release siRNA upon light irradiation and demonstrates the importance of thoroughly characterizing photoresponsive nanosystems for applications in triggered gene regulation.
Collapse
Affiliation(s)
- Rachel S. Riley
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19711, United States
| | - Megan N. Dang
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19711, United States
| | - Margaret M. Billingsley
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19711, United States
| | - Baxter Abraham
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19711, United States
| | - Lars Gundlach
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware 19711, United States
- Department of Physics and Astronomy, University of Delaware, Newark, Delaware 19711, United States
| | - Emily S. Day
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19711, United States
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19711, United States
- Helen F. Graham Cancer Center & Research Institute, Newark, Delaware 19713, United States
| |
Collapse
|
49
|
Wu C, Chen H, Wu X, Cong X, Wang L, Wang Y, Yang Y, Li W, Sun T. The influence of tumor-induced immune dysfunction on the immune cell distribution of gold nanoparticles in vivo. Biomater Sci 2018; 5:1531-1536. [PMID: 28589972 DOI: 10.1039/c7bm00335h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Gold nanoparticles (AuNPs) have been extensively explored as a drug carrier and have been widely used to provide advanced biomedical research tools in diagnostic imaging and therapy for cancer. Although the mononuclear phagocyte system and immune system are known to play the main roles in the clearance of AuNPs during the circulation, the particle distribution within the immune cells under the condition of immune dysfunction caused by tumor growth has not been thoroughly studied. Here, the cellular distribution of Cy5 labeled AuNPs with diameters of 5, 30 and 50 nm is characterized within the immune populations of the blood, spleen and bone marrow from tumor free and tumor bearing mice using flow cytometry. Tumor-associated immune dysfunction was observed in all immune organs and cell lineages, and it changed with tumor growth. Furthermore, the particle cellular distribution significantly changed in the tumor bearing mice compared with the tumor free mice. Finally, the particle distribution in the immune cells was also different at different stages of the tumor. Overall, these results can help inform and influence future AuNP design criteria including the future applications for nanoparticle-mediated cancer therapy.
Collapse
Affiliation(s)
- Chenxi Wu
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Hongmei Chen
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Xuan Wu
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| | - Xiuxiu Cong
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| | - Li Wang
- School of Life Science, University of Science & Technology of China, Hefei, Anhui, China
| | - Yucai Wang
- School of Life Science, University of Science & Technology of China, Hefei, Anhui, China
| | - Yongguang Yang
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China and Columbia Center for Translational Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Wei Li
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Tianmeng Sun
- The First Hospital of Jilin University, Changchun, Jilin, 130021, China. and Institute of Immunology, Jilin University, Changchun, Jilin, China
| |
Collapse
|
50
|
Abstract
Although viral vectors comprise the majority of gene delivery vectors, their various safety, production, and other practical concerns have left a research gap to be addressed. The non-viral vector space encompasses a growing variety of physical and chemical methods capable of gene delivery into the nuclei of target cells. Major physical methods described in this chapter are microinjection, electroporation, and ballistic injection, magnetofection, sonoporation, optical transfection, and localized hyperthermia. Major chemical methods described in this chapter are lipofection, polyfection, gold complexation, and carbon-based methods. Combination approaches to improve transfection efficiency or reduce immunological response have shown great promise in expanding the scope of non-viral gene delivery.
Collapse
Affiliation(s)
- Chi Hong Sum
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | | | - Shirley Wong
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | | |
Collapse
|