1
|
Xu Z, Li J, Yan N, Liu X, Deng Y, Song Y. Phosphatidylserine and/or Sialic Acid Modified Liposomes Increase Uptake by Tumor-associated Macrophages and Enhance the Anti-tumor Effect. AAPS PharmSciTech 2024; 25:125. [PMID: 38834759 DOI: 10.1208/s12249-024-02837-3] [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: 01/07/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
DOX liposomes have better therapeutic effects and lower toxic side effects. The targeting ability of liposomes is one of the key factors affecting the therapeutic effect of DOX liposomes. This study developed two types of targeted liposomes. Sialic acid (SA)-modified liposomes were designed to target the highly expressed Siglec-1 receptor on tumor-associated macrophages surface. Phosphatidylserine (PS)-modified liposomes were designed to promote phagocytosis by monocyte-derived macrophages through PS apoptotic signaling. In order to assess and compare the therapeutic potential of different targeted pathways in the context of anti-tumor treatment, we compared four phosphatidylserine membrane materials (DOPS, DSPS, DPPS and DMPS) and found that liposomes prepared using DOPS as material could significantly improve the uptake ability of RAW264.7 cells for DOX liposomes. On this basis, normal DOX liposomes (CL-DOX) and SA-modified DOX liposomes (SAL-DOX), PS-modified DOX liposomes (PS-CL-DOX), SA and PS co-modified DOX liposomes (PS-SAL-DOX) were prepared. The anti-tumor cells function of each liposome on S180 and RAW264.7 in vitro was investigated, and it was found that SA on the surface of liposomes can increase the inhibitory effect. In vivo efficacy results exhibited that SAL-DOX and PS-CL-DOX were superior to other groups in terms of ability to inhibit tumor growth and tumor inhibition index, among which SAL-DOX had the best anti-tumor effect. Moreover, SAL-DOX group mice had high expression of IFN-γ as well as IL-12 factors, which could significantly inhibit mice tumor growth, improve the immune microenvironment of the tumor site, and have excellent targeted delivery potential.
Collapse
Affiliation(s)
- Zihan Xu
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Jie Li
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Na Yan
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Xinrong Liu
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yihui Deng
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yanzhi Song
- College of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.
| |
Collapse
|
2
|
Ru J, Chen Y, Tao S, Du S, Liang C, Teng Z, Gao Y. Exploring Hollow Mesoporous Silica Nanoparticles as a Nanocarrier in the Delivery of Foot-And-Mouth Disease Virus-like Particle Vaccines. ACS APPLIED BIO MATERIALS 2024; 7:1064-1072. [PMID: 38286026 DOI: 10.1021/acsabm.3c01015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
Virus-like particle (VLP) vaccine is considered to be the most promising candidate alternative to the traditional inactivated vaccine for foot-and-mouth disease (FMD). To elicit a desired immune response, hollow mesoporous silica nanoparticles (HMSNs) have been synthesized and utilized as a nanocarrier for FMD VLP vaccine delivery. The as-prepared HMSNs displayed a relatively small particle size (∼260 nm), large cavity (∼150 nm), and thin wall (∼55 nm). The inherent structural superiorities make them ideal nanocarriers for the FMD VLP vaccine, which exhibited good biocompatibility, great protein-loading capacity, high antibody-response level, and protective efficiency, even comparable to commercial adjuvant ISA 206. All the results suggested that HMSNs may be a valid nanocarrier in VLP-based vaccines.
Collapse
Affiliation(s)
- Jiaxi Ru
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 325035, P. R. China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, P. R. China
| | - Yu Chen
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 325035, P. R. China
| | - Siyi Tao
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 325035, P. R. China
| | - Shaobo Du
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, P. R. China
| | - Chao Liang
- Institute for Advanced Research, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 325035, P. R. China
| | - Zhidong Teng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, P. R. China
| | - Yuan Gao
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, P. R. China
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, P. R. China
| |
Collapse
|
3
|
Yazdan M, Naghib SM, Mozafari MR. Liposomal Nano-Based Drug Delivery Systems for Breast Cancer Therapy: Recent Advances and Progresses. Anticancer Agents Med Chem 2024; 24:896-915. [PMID: 38529608 DOI: 10.2174/0118715206293653240322041047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/21/2024] [Accepted: 02/27/2024] [Indexed: 03/27/2024]
Abstract
Breast cancer is a highly prevalent disease on a global scale, with a 30% incidence rate among women and a 14% mortality rate. Developing countries bear a disproportionate share of the disease burden, while countries with greater technological advancements exhibit a higher incidence. A mere 7% of women under the age of 40 are diagnosed with breast cancer, and the prevalence of this ailment is significantly diminished among those aged 35 and younger. Chemotherapy, radiation therapy, and surgical intervention comprise the treatment protocol. However, the ongoing quest for a definitive cure for breast cancer continues. The propensity for cancer stem cells to metastasize and resistance to treatment constitute their Achilles' heel. The advancement of drug delivery techniques that target cancer cells specifically holds significant promise in terms of facilitating timely detection and effective intervention. Novel approaches to pharmaceutical delivery, including nanostructures and liposomes, may bring about substantial changes in the way breast cancer is managed. These systems offer a multitude of advantages, such as heightened bioavailability, enhanced solubility, targeted tumor destruction, and diminished adverse effects. The application of nano-drug delivery systems to administer anti-breast cancer medications is a significant subject of research. This article delves into the domain of breast cancer, conventional treatment methods, the incorporation of nanotechnology into managerial tactics, and strategic approaches aimed at tackling the disease at its core.
Collapse
Affiliation(s)
- Mostafa Yazdan
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - M R Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, VIC 3168, Australia
| |
Collapse
|
4
|
Min SH, Lei W, Jun CJ, Yan ZS, Guang YX, Tong Z, Yong ZP, Hui LZ, Xing H. Design strategy and research progress of multifunctional nanoparticles in lung cancer therapy. Expert Opin Investig Drugs 2023; 32:723-739. [PMID: 37668152 DOI: 10.1080/13543784.2023.2254683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/01/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
INTRODUCTION Lung cancer is one of the cancer types with the highest mortality rate, exploring a more effective treatment modality that improves therapeutic efficacy while mitigating side effects is now an urgent requirement. Designing multifunctional nanoparticles can be used to overcome the limitations of drugs and conventional drug delivery systems. Nanotechnology has been widely researched, and through different needs, suitable nanocarriers can be selected to load anti-cancer drugs to improve the therapeutic effect. It is foreseeable that with the rapid development of nanotechnology, more and more lung cancer patients will benefit from nanotechnology. This paper reviews the merits of various multifunctional nanoparticles in the treatment of lung cancer to provide novel ideas for lung cancer treatment. AREAS COVERED This review focuses on summarizing various nanoparticles for targeted lung cancer therapy and their advantages and disadvantages, using nanoparticles loaded with anti-cancer drugs, delivered to lung cancer sites, enhancing drug half-life, improving anti-cancer drug efficacy and reducing side effects. EXPERT OPINION The delivery mode of nanoparticles with superior pharmacokinetic properties in the in vivo circulation enhances the half-life of the drug, and provides tissue-targeted selectivity and the ability to overcome biological barriers, bringing a revolution in the field of oncology.
Collapse
Affiliation(s)
- Shen Hui Min
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Lei
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chen Jia Jun
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhang Shao Yan
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Xu Guang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhang Tong
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng Pei Yong
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Zhen Hui
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huang Xing
- Institute of Respiratory Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Hybrid Magnetic Lipid-Based Nanoparticles for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15030751. [PMID: 36986612 PMCID: PMC10058222 DOI: 10.3390/pharmaceutics15030751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/05/2023] Open
Abstract
Cancer is one of the major public health problems worldwide. Despite the advances in cancer therapy, it remains a challenge due to the low specificity of treatment and the development of multidrug resistance mechanisms. To overcome these drawbacks, several drug delivery nanosystems have been investigated, among them, magnetic nanoparticles (MNP), especially superparamagnetic iron oxide nanoparticles (SPION), which have been applied for treating cancer. MNPs have the ability to be guided to the tumor microenvironment through an external applied magnetic field. Furthermore, in the presence of an alternating magnetic field (AMF) this nanocarrier can transform electromagnetic energy in heat (above 42 °C) through Néel and Brown relaxation, which makes it applicable for hyperthermia treatment. However, the low chemical and physical stability of MNPs makes their coating necessary. Thus, lipid-based nanoparticles, especially liposomes, have been used to encapsulate MNPs to improve their stability and enable their use as a cancer treatment. This review addresses the main features that make MNPs applicable for treating cancer and the most recent research in the nanomedicine field using hybrid magnetic lipid-based nanoparticles for this purpose.
Collapse
|
6
|
Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm (Beijing) 2023; 4:e187. [PMID: 36654533 PMCID: PMC9834710 DOI: 10.1002/mco2.187] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2023] Open
Abstract
Cancer is a complex disease associated with a combination of abnormal physiological process and exhibiting dysfunctions in multiple systems. To provide effective treatment and diagnosis for cancer, current treatment strategies simultaneously focus on various tumor targets. Based on the rapid development of nanotechnology, nanocarriers have been shown to exhibit excellent potential for cancer therapy. Compared with nanoparticles with single functions, multifunctional nanoparticles are believed to be more aggressive and potent in the context of tumor targeting. However, the development of multifunctional nanoparticles is not simply an upgraded version of the original function, but involves a sophisticated system with a proper backbone, optimized modification sites, simple preparation method, and efficient function integration. Despite this, many well-designed multifunctional nanoparticles with promising therapeutic potential have emerged recently. Here, to give a detailed understanding and analyzation of the currently developed multifunctional nanoparticles, their platform structures with organic or inorganic backbones were systemically generalized. We emphasized on the functionalization and modification strategies, which provide additional functions to the nanoparticle. We also discussed the application combination strategies that were involved in the development of nanoformulations with functional crosstalk. This review thus provides an overview of the construction strategies and application advances of multifunctional nanoparticles.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Xingmei Duan
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuan ProvinceChina
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
7
|
Hashemi M, Ghadyani F, Hasani S, Olyaee Y, Raei B, Khodadadi M, Ziyarani MF, Basti FA, Tavakolpournegari A, Matinahmadi A, Salimimoghadam S, Aref AR, Taheriazam A, Entezari M, Ertas YN. Nanoliposomes for doxorubicin delivery: Reversing drug resistance, stimuli-responsive carriers and clinical translation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
8
|
Toro-Córdova A, Llaguno-Munive M, Jurado R, Garcia-Lopez P. The Therapeutic Potential of Chemo/Thermotherapy with Magnetoliposomes for Cancer Treatment. Pharmaceutics 2022; 14:2443. [PMID: 36432634 PMCID: PMC9697689 DOI: 10.3390/pharmaceutics14112443] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer represents a very grave and quickly growing public health problem worldwide. Despite the breakthroughs in treatment and early detection of the disease, an increase is projected in the incidence rate and mortality during the next 30 years. Thus, it is important to develop new treatment strategies and diagnostic tools. One alternative is magnetic hyperthermia, a therapeutic approach that has shown promising results, both as monotherapy and in combination with chemo- and radiotherapy. However, there are still certain limitations and questions with respect to the safety of the systemic administration of magnetic nanoparticles. To deal with these issues, magnetoliposomes were conceived as a new generation of liposomes that incorporate superparamagnetic nanoparticles and oncological pharmaceuticals within their structure. They have the advantage of targeted and selective drug delivery to the diseased organs and tissues. Some of them can avoid the immune response of the host. When exposed to a magnetic field of alternating current, magnetoliposomes produce hyperthermia, which acts synergistically with the released drug. The aim of the present review is to describe the most recent advances in the use of magnetoliposomes and point out what research remains to be done for their application to chemo-thermal therapy in cancer patients.
Collapse
Affiliation(s)
- Alfonso Toro-Córdova
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
- Departamento de Formulación de Vacunas de mRNA, CerTest Biotec S.L., 50840 Zaragoza, Spain
| | - Monserrat Llaguno-Munive
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
- Laboratorio de Física Médica, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| | - Rafael Jurado
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| | - Patricia Garcia-Lopez
- Laboratorio de Fármaco-Oncología, Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Cd, Mexico City 14080, Mexico
| |
Collapse
|
9
|
Pu XQ, Ju XJ, Liu WY, Liu YQ, Li XJ, Li Y, Xie R, Wang W, Liu Z, Chu LY. Stimulus-Responsive Nanoparticle-Integrated Dissolving Microneedles for Synergetic Chemo-Photothermal Therapy of Superficial Skin Tumors. Ind Eng Chem Res 2022. [DOI: 10.1021/acs.iecr.2c00831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Xing-Qun Pu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Xiao-Jie Ju
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Wen-Ying Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Yu-Qiong Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Xin-Jiao Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Yao Li
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Rui Xie
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Wei Wang
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Zhuang Liu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| | - Liang-Yin Chu
- School of Chemical Engineering, Sichuan University, Chengdu 610065, Sichuan, China
- State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, Sichuan, China
| |
Collapse
|
10
|
Fan X, Wang Y, Li B, Shen C, Sun Z, Zhan Y, Zhang Y. Highly luminescent pH-responsive carbon quantum dots for cell imaging. NANOTECHNOLOGY 2022; 33:265002. [PMID: 35299160 DOI: 10.1088/1361-6528/ac5ee5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/16/2022] [Indexed: 06/14/2023]
Abstract
Carbon quantum dots (CDs) have attracted tremendous interest owing to their idiosyncratic functions and wide-ranging applications. However, it remains a great challenge to empolder an integrated CDs combining high luminescence, biocompatibility and luminescence color tunability for bioimaging via simple approach. In this work, pH-responsive carbon quantum dots (Si-CDs) with high luminescence (quantum yield = 74.8%) were fabricated by one-step hydrothermal method using (3-mercaptopropyl) triethoxysilane (KH-580) as modifier for the first time. The optical properties of the as-prepared Si-CDs can be controlled from obvious green-blue-violet transformation by altering the pH. More importantly, the change is reversible and repeatable. In addition, the Si-CDs have good biocompatibility and chemically inertin vitrocell system simulation. Such non-toxic, environmental friendly, low-cost, inert CDs materials are promising candidates for biomedical and pH-sensitive sensors.
Collapse
Affiliation(s)
- Xiaohui Fan
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, People's Republic of China
| | - Yang Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, People's Republic of China
| | - Bo Li
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, People's Republic of China
| | - Chang Shen
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, People's Republic of China
| | - Zhengguang Sun
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, People's Republic of China
| | - Yuan Zhan
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, People's Republic of China
| | - Yuhong Zhang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, Ministry of Education Key Laboratory for the Synthesis and Application of Organic Functional Molecules, College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, People's Republic of China
| |
Collapse
|
11
|
Wu L, Wen W, Wang X, Huang D, Cao J, Qi X, Shen S. Ultrasmall iron oxide nanoparticles cause significant toxicity by specifically inducing acute oxidative stress to multiple organs. Part Fibre Toxicol 2022; 19:24. [PMID: 35351185 PMCID: PMC8962100 DOI: 10.1186/s12989-022-00465-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/17/2022] [Indexed: 01/21/2023] Open
Abstract
Background Iron oxide nanoparticles have been approved by food and drug administration for clinical application as magnetic resonance imaging (MRI) and are considered to be a biocompatible material. Large iron oxide nanoparticles are usually used as transversal (T2) contrast agents to exhibit dark contrast in MRI. In contrast, ultrasmall iron oxide nanoparticles (USPIONs) (several nanometers) showed remarkable advantage in longitudinal (T1)-weighted MRI due to the brighten effect. The study of the toxicity mainly focuses on particles with size of tens to hundreds of nanometers, while little is known about the toxicity of USPIONs. Results We fabricated Fe3O4 nanoparticles with diameters of 2.3, 4.2, and 9.3 nm and evaluated their toxicity in mice by intravenous injection. The results indicate that ultrasmall iron oxide nanoparticles with small size (2.3 and 4.2 nm) were highly toxic and were lethal at a dosage of 100 mg/kg. In contrast, no obvious toxicity was observed for iron oxide nanoparticles with size of 9.3 nm. The toxicity of small nanoparticles (2.3 and 4.2 nm) could be reduced when the total dose was split into 4 doses with each interval for 5 min. To study the toxicology, we synthesized different-sized SiO2 and gold nanoparticles. No significant toxicity was observed for ultrasmall SiO2 and gold nanoparticles in the mice. Hence, the toxicity of the ultrasmall Fe3O4 nanoparticles should be attributed to both the iron element and size. In the in vitro experiments, all the ultrasmall nanoparticles (< 5 nm) of Fe3O4, SiO2, and gold induced the generation of the reactive oxygen species (ROS) efficiently, while no obvious ROS was observed in larger nanoparticles groups. However, the ·OH was only detected in Fe3O4 group instead of SiO2 and gold groups. After intravenous injection, significantly elevated ·OH level was observed in heart, serum, and multiple organs. Among these organs, heart showed highest ·OH level due to the high distribution of ultrasmall Fe3O4 nanoparticles, leading to the acute cardiac failure and death. Conclusion Ultrasmall Fe3O4 nanoparticles (2.3 and 4.2 nm) showed high toxicity in vivo due to the distinctive capability in inducing the generation of ·OH in multiple organs, especially in heart. The toxicity was related to both the iron element and size. These findings provide novel insight into the toxicology of ultrasmall Fe3O4 nanoparticles, and also highlight the need of comprehensive evaluation for their clinic application. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12989-022-00465-y.
Collapse
|
12
|
Zhang L, Guan X, Xiao X, Chai Y, Chen Z, Zhou G, Fan Y. Near-infrared triggered injectable ferrimagnetic chitosan thermosensitive hydrogel for photo hyperthermia and precisely controlled drug release in tumor ablation. Eur Polym J 2022. [DOI: 10.1016/j.eurpolymj.2021.110879] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
13
|
Józefczak A, Kaczmarek K, Bielas R. Magnetic mediators for ultrasound theranostics. Theranostics 2021; 11:10091-10113. [PMID: 34815806 PMCID: PMC8581415 DOI: 10.7150/thno.62218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/02/2021] [Indexed: 12/11/2022] Open
Abstract
The theranostics paradigm is based on the concept of combining therapeutic and diagnostic modalities into one platform to improve the effectiveness of treatment. Combinations of multiple modalities provide numerous medical advantages and are enabled by nano- and micron-sized mediators. Here we review recent advancements in the field of ultrasound theranostics and the use of magnetic materials as mediators. Several subdisciplines are described in detail, including controlled drug delivery and release, ultrasound hyperthermia, magneto-ultrasonic heating, sonodynamic therapy, magnetoacoustic imaging, ultrasonic wave generation by magnetic fields, and ultrasound tomography. The continuous progress and improvement in theranostic materials, methods, and physical computing models have created undeniable possibilities for the development of new approaches. We discuss the prospects of ultrasound theranostics and possible expansions of other studies to the theranostic context.
Collapse
Affiliation(s)
- Arkadiusz Józefczak
- Chair of Acoustics, Faculty of Physics, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 2, 61-614 Poznań, Poland
| | - Katarzyna Kaczmarek
- Department of Biomedical Engineering, Faculty of Engineering, University of Strathclyde, Wolfson Centre, 106 Rottenrow, Glasgow, United Kingdom
| | - Rafał Bielas
- Chair of Acoustics, Faculty of Physics, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 2, 61-614 Poznań, Poland
| |
Collapse
|
14
|
Karimkhah F, Elhamifar D, Shaker M. Ag 2CO 3 containing magnetic nanocomposite as a powerful and recoverable catalyst for Knoevenagel condensation. Sci Rep 2021; 11:18736. [PMID: 34548589 PMCID: PMC8455631 DOI: 10.1038/s41598-021-98287-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/30/2021] [Indexed: 02/08/2023] Open
Abstract
In this paper, the synthesis, characterization and catalytic application of a novel magnetic silica-supported Ag2CO3 (MS/Ag2CO3) with core-shell structure are developed. The MS/Ag2CO3 nanocomposite was prepared through chemical modification of magnetic MS nanoparticles with AgNO3 under alkaline conditions. The structure, chemical composition and magnetic properties of MS/Ag2CO3 were investigated by using VSM, PXRD, FT-IR, EDX and SEM techniques. The MS/Ag2CO3 nanocomposite was used as an effective catalyst for the Knoevenagel condensation under solvent-free conditions at 60 °C in an ultrasonic bath. The recovery and leaching tests were performed to study the nature of the MS/Ag2CO3 catalyst under applied conditions.
Collapse
Affiliation(s)
- Fatemeh Karimkhah
- Department of Chemistry, Yasouj University, 75918-74831, Yasouj, Iran
| | - Dawood Elhamifar
- Department of Chemistry, Yasouj University, 75918-74831, Yasouj, Iran.
| | - Masoumeh Shaker
- Department of Chemistry, Yasouj University, 75918-74831, Yasouj, Iran
| |
Collapse
|
15
|
Tang G, He J, Liu J, Yan X, Fan K. Nanozyme for tumor therapy: Surface modification matters. EXPLORATION (BEIJING, CHINA) 2021; 1:75-89. [PMID: 37366468 PMCID: PMC10291575 DOI: 10.1002/exp.20210005] [Citation(s) in RCA: 171] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/25/2021] [Indexed: 06/28/2023]
Abstract
As the next generation of artificial enzymes, nanozymes have shown unique properties compared to its natural counterparts, such as stability in harsh environment, low cost, and ease of production and modification, paving the way for its biomedical applications. Among them, tumor catalytic therapy mediated by the generation of reactive oxygen species (ROS) has made great progress mainly from the peroxidase-like activity of nanozymes. Fe3O4 nanozymes, the earliest type of nanomaterial discovered to possess peroxidase-like activity, has consequently received wide attention for tumor therapy due to its ROS generation ability and tumor cell killing ability. However, inconsistent results of cytotoxicity were observed between different reports, and some even showed the scavenging of ROS in some cases. By collectively studying these inconsistent outcomes, we raise the question whether surface modification of Fe3O4 nanozymes, either through affecting peroxidase activity or by affecting the biodistribution and intracellular fate, play an important role in its therapeutic effects. This review will go over the fundamental catalytic mechanisms of Fe3O4 nanozymes and recent advances in tumor catalytic therapy, and discuss the importance of surface modification. Employing Fe3O4 nanozymes as an example, we hope to provide an outlook on the improvement of nanozyme-based antitumor activity.
Collapse
Affiliation(s)
- Guoheng Tang
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesBeijing101408P. R. China
| | - Jiuyang He
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijing100101P. R. China
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for NanotechnologyUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Xiyun Yan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesBeijing101408P. R. China
- Nanozyme Medical Center, School of Basic Medical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of BiophysicsChinese Academy of SciencesBeijing100101P. R. China
- University of Chinese Academy of SciencesBeijing101408P. R. China
- Nanozyme Medical Center, School of Basic Medical SciencesZhengzhou UniversityZhengzhou450001P. R. China
| |
Collapse
|
16
|
Triple negative breast cancer and non-small cell lung cancer: Clinical challenges and nano-formulation approaches. J Control Release 2021; 337:27-58. [PMID: 34273417 DOI: 10.1016/j.jconrel.2021.07.014] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 02/06/2023]
Abstract
Triple negative breast cancer (TNBC) and non-small cell lung cancer (NSCLC) are amongst the most aggressive forms of solid tumors. TNBC is highlighted by absence of genetic components of progesterone receptor, HER2/neu and estrogen receptor in breast cancer. NSCLC is characterized by integration of malignant carcinoma into respiratory system. Both cancers are associated with poor median and overall survival rates with low progression free survival with high incidences of relapse. These cancers are characterized by tumor heterogeneity, genetic mutations, generation of cancer-stem cells, immune-resistance and chemoresistance. Further, these neoplasms have been reported for tumor cross-talk into second primary cancers for each other. Current chemotherapeutic regimens include usage of multiple agents in tandem to affect tumor cells through multiple mechanisms with various such combinations being clinically tested. However, lack of controlled delivery and effective temporospatial presence of chemotherapeutics has resulted in suboptimal therapeutic response. Consequently, passive targeted albumin bound paclitaxel and PEGylated liposomal doxorubicin have been clinically used and tested with newer drugs for improved therapeutic efficacy in these cancers. Active targeting of nanocarriers against surface overexpressed proteins in both neoplasms have been explored. However, use of single agent nanoparticulate formulations against both cancers have failed to elicit desired outcomes. This review aims to identify clinical unmet need in these cancers while establishing a correlation with tested nano-formulation approaches and issues with preclinical to clinical translation. Lipid and polymer-based drug-drug and drug-gene combinatorial nanocarriers delivering multiple chemotherapeutics simultaneously to desired site of action have been detailed. Finally, emerging opportunities such as pharmacological targets (immune check point and epigentic modulators) as well as gene-based modulation (siRNA/CRISPR/Cas9) and the nano-formulation challenges for effective treatment of both cancers have been explored.
Collapse
|
17
|
Dimitriou P, Li J, Tornillo G, McCloy T, Barrow D. Droplet Microfluidics for Tumor Drug-Related Studies and Programmable Artificial Cells. GLOBAL CHALLENGES (HOBOKEN, NJ) 2021; 5:2000123. [PMID: 34267927 PMCID: PMC8272004 DOI: 10.1002/gch2.202000123] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/19/2021] [Indexed: 05/11/2023]
Abstract
Anticancer drug development is a crucial step toward cancer treatment, that requires realistic predictions of malignant tissue development and sophisticated drug delivery. Tumors often acquire drug resistance and drug efficacy, hence cannot be accurately predicted in 2D tumor cell cultures. On the other hand, 3D cultures, including multicellular tumor spheroids (MCTSs), mimic the in vivo cellular arrangement and provide robust platforms for drug testing when grown in hydrogels with characteristics similar to the living body. Microparticles and liposomes are considered smart drug delivery vehicles, are able to target cancerous tissue, and can release entrapped drugs on demand. Microfluidics serve as a high-throughput tool for reproducible, flexible, and automated production of droplet-based microscale constructs, tailored to the desired final application. In this review, it is described how natural hydrogels in combination with droplet microfluidics can generate MCTSs, and the use of microfluidics to produce tumor targeting microparticles and liposomes. One of the highlights of the review documents the use of the bottom-up construction methodologies of synthetic biology for the formation of artificial cellular assemblies, which may additionally incorporate both target cancer cells and prospective drug candidates, as an integrated "droplet incubator" drug assay platform.
Collapse
Affiliation(s)
- Pantelitsa Dimitriou
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Jin Li
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Giusy Tornillo
- Hadyn Ellis BuildingCardiff UniversityMaindy RoadCardiffCF24 4HQUK
| | - Thomas McCloy
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - David Barrow
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| |
Collapse
|
18
|
Ferri-Liposomes: Preformulation and Selective Cytotoxicity against A549 Lung Cancer Cells. Pharmaceutics 2021; 13:pharmaceutics13050712. [PMID: 34068129 PMCID: PMC8152733 DOI: 10.3390/pharmaceutics13050712] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
Liposomes have become successful nanostructured systems used in clinical practices. These vesicles are able to carry important drug loadings with noteworthy stability. The aim of this work was to develop iron oxide-loaded stealth liposomes as a prospective alternative for the treatment of lung cancer. In this study, citric acid iron oxide nanoparticles (IONPs-Ac) were synthesized and encapsulated in stealth liposomes. Their cytotoxicity and selectivity against lung tumor cells were assessed. Stealth liposomal vesicles, with relevant content of IONPs-Ac, named ferri-liposomes (SL-IONPs-Ac), were produced with an average size of 200 nm. They displayed important cytotoxicity in a human lung cancer cells model (A549 cells), even at low concentrations, whereas free IONPs-Ac displayed adequate biocompatibility. Nevertheless, the treatment at the same concentration of ferri-liposomes against HEK-293 cells, a normal human cell lineage, was not significantly cytotoxic, revealing a probable lung tumor selectiveness of the fabricated formulation. Furthermore, from the flow cytometry studies, it was possible to infer that ferri-liposomes were able to induce A549 tumor cells death through apoptosis/ferroptosis processes, evidenced by a significant reduction of the mitochondrial membrane potential.
Collapse
|
19
|
Veloso SRS, Andrade RGD, Castanheira EMS. Magnetoliposomes: recent advances in the field of controlled drug delivery. Expert Opin Drug Deliv 2021; 18:1323-1334. [PMID: 33836636 DOI: 10.1080/17425247.2021.1915983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Magnetoliposomes have gained increasing attention as delivery systems, as they surpass many limitations associated with liposomes. The combination with magnetic nanoparticles provides a means for development of multimodal and multifunctional theranostic agents that enable on-demand drug release and real-time monitoring of therapy. AREAS COVERED Recently, several magnetoliposome structures have been reported to ensure efficient transport and delivery of therapeutics, while improving magnetic properties. Besides, novel techniques have been introduced to improve on-demand release, as well as to achieve sequential release of different therapeutic agents. This review presents the major types and methods of preparation of magnetoliposomes, and discusses recent strategies in the trigger of drug release, development of theranostic formulations, and delivery of drugs and biological entities. EXPERT OPINION Despite significant advances in efficient drug delivery, current literature lacks an assessment of formulations as theranostic agents and complementary techniques to optimize thermotherapy efficiency. Plasmonic magnetoliposomes are highly promising multimodal and multifunctional systems, providing the required design versatility to optimize theranostic capabilities. Further, photodynamic therapy and delivery of proteins/genes can be improved with a deeper research on the employed magnetic material and associated toxicity. A scale-up procedure is also lacking in recent research, which is limiting their translation to clinical use.
Collapse
Affiliation(s)
- Sérgio R S Veloso
- Physics Center of Minho and Porto Universities (CF-UM-UP), University of Minho, Campus de Gualtar, Braga, Portugal
| | - Raquel G D Andrade
- Physics Center of Minho and Porto Universities (CF-UM-UP), University of Minho, Campus de Gualtar, Braga, Portugal
| | - Elisabete M S Castanheira
- Physics Center of Minho and Porto Universities (CF-UM-UP), University of Minho, Campus de Gualtar, Braga, Portugal
| |
Collapse
|
20
|
Yuba E, Sugahara Y, Yoshizaki Y, Shimizu T, Kasai M, Udaka K, Kono K. Carboxylated polyamidoamine dendron-bearing lipid-based assemblies for precise control of intracellular fate of cargo and induction of antigen-specific immune responses. Biomater Sci 2021; 9:3076-3089. [PMID: 33681873 DOI: 10.1039/d0bm01813a] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
For the establishment of advanced medicines such as cancer immunotherapy, high performance carriers that precisely deliver biologically active molecules must be developed to target organelles of the cells and to release their contents there. From the viewpoint of antigen delivery, endosomes are important target organelles because they contain immune-response-related receptors and proteins of various types. To obtain carriers for precision endosome delivery, a novel type of polyamidoamine dendron-based lipid having pH-sensitive terminal groups was synthesized for this study. Liposomes were prepared using these pH-sensitive dendron-based lipids and egg yolk phosphatidylcholine. Their pH-responsive properties and performance as an endosome delivery carrier were investigated. pH-Sensitive dendron lipid-based liposomes retained water-soluble molecules at neutral pH but released them under weakly acidic conditions. Particularly, liposomes containing CHexDL-G1U exhibited highly sensitive properties responding to very weakly acidic pH. These dendron lipid-based liposomes released the contents specifically in the endosome. The timing of content release can be controlled by selecting pH-sensitive dendron lipids for liposome preparation. Significant tumor regression was induced in tumor-bearing mice by the administration of CHexDL-G1U-modified liposomes containing the model antigenic protein. Furthermore, CHexDL-G1U-modified liposomes induced WT1 tumor antigenic peptide-specific helper T cell proliferation. The results demonstrate that dendron lipid-based liposomes are useful as a potent vaccine for cancer immunotherapy.
Collapse
Affiliation(s)
- Eiji Yuba
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Yoshikatsu Sugahara
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Yuta Yoshizaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| | - Takeyuki Shimizu
- Department of Immunology, School of Medicine, Kochi University, Nankoku, Kochi 7838505, Japan
| | - Michiyuki Kasai
- Department of Immunology, School of Medicine, Kochi University, Nankoku, Kochi 7838505, Japan
| | - Keiko Udaka
- Department of Immunology, School of Medicine, Kochi University, Nankoku, Kochi 7838505, Japan
| | - Kenji Kono
- Department of Applied Chemistry, Graduate School of Engineering, Osaka Prefecture University, 1-1 Gakuen-cho, Naka-ku, Sakai, Osaka 5998531, Japan.
| |
Collapse
|
21
|
Anik MI, Hossain MK, Hossain I, Mahfuz AMUB, Rahman MT, Ahmed I. Recent progress of magnetic nanoparticles in biomedical applications: A review. NANO SELECT 2021. [DOI: 10.1002/nano.202000162] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Muzahidul I. Anik
- Chemical Engineering University of Rhode Island Kingston Rhode Island 02881 USA
| | - M. Khalid Hossain
- Interdisciplinary Graduate School of Engineering Science Kyushu University Fukuoka 816–8580 Japan
- Atomic Energy Research Establishment Bangladesh Atomic Energy Commission Dhaka 1349 Bangladesh
| | - Imran Hossain
- Institute for Micromanufacturing Louisiana Tech University Ruston Louisiana 71270 USA
| | - A. M. U. B. Mahfuz
- Biotechnology and Genetic Engineering University of Development Alternative Dhaka 1209 Bangladesh
| | - M. Tayebur Rahman
- Materials Science and Engineering University of Rajshahi Rajshahi 6205 Bangladesh
| | - Isteaque Ahmed
- Chemical Engineering University of Cincinnati Cincinnati Ohio 45221 USA
| |
Collapse
|
22
|
Wang Q, Xiao J, Su Y, Huang J, Li J, Qiu L, Zhan M, He X, Yuan W, Li Y. Fabrication of thermoresponsive magnetic micelles from amphiphilic poly(phenyl isocyanide) and Fe3O4 nanoparticles for controlled drug release and synergistic thermochemotherapy. Polym Chem 2021. [DOI: 10.1039/d1py00022e] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The drug-loaded micelles self-assembled from co-poly(phenyl isocyanide), Fe3O4 and DOX demonstrated thermoresponsiveness and magnetic hyperthermia for synergistic thermochemotherapy.
Collapse
|
23
|
Alijani H, Noori A, Faridi N, Bathaie S, Mousavi MF. Aptamer-functionalized Fe3O4@MOF nanocarrier for targeted drug delivery and fluorescence imaging of the triple-negative MDA-MB-231 breast cancer cells. J SOLID STATE CHEM 2020. [DOI: 10.1016/j.jssc.2020.121680] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
24
|
Enhanced In Vitro Magnetic Cell Targeting of Doxorubicin-Loaded Magnetic Liposomes for Localized Cancer Therapy. NANOMATERIALS 2020; 10:nano10112104. [PMID: 33114052 PMCID: PMC7690690 DOI: 10.3390/nano10112104] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022]
Abstract
The lack of efficient targeting strategies poses significant limitations on the effectiveness of chemotherapeutic treatments. This issue also affects drug-loaded nanocarriers, reducing nanoparticles cancer cell uptake. We report on the fabrication and in vitro characterization of doxorubicin-loaded magnetic liposomes for localized treatment of liver malignancies. Colloidal stability, superparamagnetic behavior and efficient drug loading of our formulation were demonstrated. The application of an external magnetic field guaranteed enhanced nanocarriers cell uptake under cell medium flow in correspondence of a specific area, as we reported through in vitro investigation. A numerical model was used to validate experimental data of magnetic targeting, proving the possibility of accurately describing the targeting strategy and predict liposomes accumulation under different environmental conditions. Finally, in vitro studies on HepG2 cancer cells confirmed the cytotoxicity of drug-loaded magnetic liposomes, with cell viability reduction of about 50% and 80% after 24 h and 72 h of incubation, respectively. Conversely, plain nanocarriers showed no anti-proliferative effects, confirming the formulation safety. Overall, these results demonstrated significant targeting efficiency and anticancer activity of our nanocarriers and superparamagnetic nanoparticles entrapment could envision the theranostic potential of the formulation. The proposed magnetic targeting study could represent a valid tool for pre-clinical investigation regarding the effectiveness of magnetic drug targeting.
Collapse
|
25
|
Ghosh S, Lalani R, Maiti K, Banerjee S, Bhatt H, Bobde YS, Patel V, Biswas S, Bhowmick S, Misra A. Synergistic co-loading of vincristine improved chemotherapeutic potential of pegylated liposomal doxorubicin against triple negative breast cancer and non-small cell lung cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 31:102320. [PMID: 33075540 DOI: 10.1016/j.nano.2020.102320] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/06/2020] [Accepted: 10/06/2020] [Indexed: 11/15/2022]
Abstract
The current work aims to explore the biological characteristics of vincristine synergistic co-loading into pegylated liposomal doxorubicin in non-indicated modalities of non-small cell lung cancer (NSCLC) and triple negative breast cancer (TNBC). The combinatorial liposome prepared by active co-loading of the drugs against modified ammonium ion gradient exhibited 95% encapsulation of both drugs. The cellular uptake studies using confocal microscopy and flow cytometry showed significantly increased uptake of dual drug formulation as against liposomal doxorubicin. The co-loaded liposome formulation had significantly increased cell cycle arrest in G2/M phase with subsequent apoptosis and reduced cell viability in both tumor cell lines than doxorubicin liposome. This carrier exhibited similar acute toxicity, pharmacokinetic and tissue distribution profiles with significant increase in tumor regression as compared to liposomal doxorubicin. These results indicate that co-encapsulation of vincristine into clinically used pegylated liposomal doxorubicin significantly improved in-vitro and in-vivo therapeutic efficacy against NSCLC and TNBC.
Collapse
Affiliation(s)
- Saikat Ghosh
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India
| | - Rohan Lalani
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Kuntal Maiti
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India
| | - Shubhadeep Banerjee
- Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India
| | - Himanshu Bhatt
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana, India
| | - Yamini Shankar Bobde
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana, India
| | - Vivek Patel
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Medchal, Hyderabad, Telangana, India
| | - Subhas Bhowmick
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Formulation Development Department - Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat, India.
| | - Ambikanandan Misra
- Department of Pharmaceutics, Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, India; Pharmaceutical Research, Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS University, Mumbai, Maharashtra, India.
| |
Collapse
|
26
|
Fathi M, Abdolahinia ED, Barar J, Omidi Y. Smart stimuli-responsive biopolymeric nanomedicines for targeted therapy of solid tumors. Nanomedicine (Lond) 2020; 15:2171-2200. [DOI: 10.2217/nnm-2020-0146] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Solid tumors form a permissive microenvironment with irregular features, including high pressured tumor interstitial fluid with acidic pH, co-adaptation of cancer cells with other cells like the immune system cells, abnormal metabolism and anomalous overexpression of various pieces of molecular machinery. The functional expressions of several oncomarkers in different solid tumors have led to the development of targeted drug-delivery systems (DDSs). As a new class of DDSs, stimuli-responsive nanomedicines (SRNMs) have been developed using advanced nanobiomaterials such as biopolymers that show excellent biocompatibility with low inherent immunogenicity. In this review, we aim to overview different types of SRNMs, present deep insights into the stimuli-responsive biopolymers and discuss the most up-to-date progress in the design and development of SRNMs used as advanced DDSs for targeted therapy of cancer.
Collapse
Affiliation(s)
- Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
27
|
In Vivo Assessment of Thermosensitive Liposomes for the Treatment of Port Wine Stains by Antifibrinolytic Site-Specific Pharmaco-Laser Therapy. Pharmaceutics 2020; 12:pharmaceutics12060591. [PMID: 32630457 PMCID: PMC7356038 DOI: 10.3390/pharmaceutics12060591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 01/10/2023] Open
Abstract
Antifibrinolytic site-specific pharmaco-laser therapy (SSPLT) is an experimental treatment modality for refractory port wine stains (PWS). Conceptually, antifibrinolytic drugs encapsulated in thermosensitive liposomes are delivered to thrombi that form in semi-photocoagulated PWS blood vessels after conventional laser treatment. Local release of antifibrinolytics is induced by mild hyperthermia, resulting in hyperthrombosis and complete occlusion of the target blood vessel (clinical endpoint). In this study, 20 thermosensitive liposomal formulations containing tranexamic acid (TA) were assayed for physicochemical properties, TA:lipid ratio, encapsulation efficiency, and endovesicular TA concentration. Two candidate formulations (DPPC:DSPE-PEG, DPPC:MPPC:DSPE-PEG) were selected based on optimal properties and analyzed for heat-induced TA release at body temperature (T), phase transition temperature (Tm), and at T > Tm. The effect of plasma on liposomal stability at 37 °C was determined, and the association of liposomes with platelets was examined by flow cytometry. The accumulation of PEGylated phosphocholine liposomes in laser-induced thrombi was investigated in a hamster dorsal skinfold model and intravital fluorescence microscopy. Both formulations did not release TA at 37 °C. Near-complete TA release was achieved at Tm within 2.0–2.5 min of heating, which was accelerated at T > Tm. Plasma exerted a stabilizing effect on both formulations. Liposomes showed mild association with platelets. Despite positive in vitro results, fluorescently labeled liposomes did not sufficiently accumulate in laser-induced thrombi in hamsters to warrant their use in antifibrinolytic SSPLT, which can be solved by coupling thrombus-targeting ligands to the liposomes.
Collapse
|
28
|
Luo X, Zhang J, Wu YP, Yang X, Kuang XP, Li WX, Li YF, He RR, Liu M. Multifunctional HNT@Fe 3O 4@PPy@DOX Nanoplatform for Effective Chemo-Photothermal Combination Therapy of Breast Cancer with MR Imaging. ACS Biomater Sci Eng 2020; 6:3361-3374. [PMID: 33463181 DOI: 10.1021/acsbiomaterials.9b01709] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Multifunctional nanoparticles for imaging and treatment in cancer are getting more and more attention recently. Herein, halloysite nanotubes (HNTs), natural clay nanotubes, are designed as multifunctional nanoplatform for targeted delivering photothermal therapy agents and chemotherapeutic drugs. Fe3O4 was anchored on the outer surfaces of HNTs and then doxorubicin (DOX) was loaded on the nanotubes. Afterward, a layer of polypyrrole (PPy), as photothermal agent, was wrapped on the tubes. The nanoplatform of HNT@Fe3O4@PPy@DOX can be guided to tumor tissue by an external magnetic field, and then performs chemo-photothermal combined therapy by 808 nm laser irradiation. HNT@Fe3O4@PPy@DOX shows the ability of T2-weighted magnetic resonance imaging, which could be considered as a promising application in magnetic targeting tumor therapy. In vitro and in vivo experiments demonstrate that HNTs nanoplatform has good biocompatibility and produces a strong antitumor effect trigged by near-infrared laser irradiation. The novel chemo-photothermal therapy nanoplatform based on HNTs may be developed as a multifunctional nanoparticle for imaging and therapy in breast cancer.
Collapse
Affiliation(s)
- Xiang Luo
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Jun Zhang
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Yan-Ping Wu
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaohan Yang
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| | - Xiu-Ping Kuang
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.,Yunnan University of Traditional Chinese Medicine, Kunming 650550, China
| | - Wei-Xi Li
- Yunnan University of Traditional Chinese Medicine, Kunming 650550, China
| | - Yi-Fang Li
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou 510632, China.,Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China.,International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Mingxian Liu
- Department of Materials Science and Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
29
|
Kostevšek N, Cheung CCL, Serša I, Kreft ME, Monaco I, Comes Franchini M, Vidmar J, Al-Jamal WT. Magneto-Liposomes as MRI Contrast Agents: A Systematic Study of Different Liposomal Formulations. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E889. [PMID: 32384645 PMCID: PMC7279489 DOI: 10.3390/nano10050889] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 04/27/2020] [Accepted: 04/30/2020] [Indexed: 12/11/2022]
Abstract
The majority of the clinically approved iron oxide nanoparticles (IO NPs) used as contrast agents for magnetic resonance imaging (MRI) have been withdrawn from the market either due to safety concerns or lack of profits. To address this challenge, liposomes have been used to prepare IO-based T2 contrast agents. We studied the influence of different phospholipids on the relaxivity (r2) values of magneto-liposomes (MLs) containing magnetic NPs in the bilayer, where a strong correlation between the bilayer fluidity and r2 is clearly shown. Embedding 5-nm IO NPs in the lipid bilayer leads to a significant improvement in their relaxivity, where r2 values range from 153 ± 5 s-1 mM-1 for DPPC/cholesterol/DSPE-PEG (96/50/4) up to 673 ± 12 s-1 mM-1 for DOPC/DSPE-PEG (96/4), compared to "free" IO NPs with an r2 value of 16 s-1 mM-1, measured at 9.4 T MRI scanner. In vitro MRI measurements, together with the ICP-MS analysis, revealed MLs as highly selective contrast agents that were preferentially taken up by cancerous T24 cells, which led to an improvement in the contrast and an easier distinction between the healthy and the cancerous cells. A careful selection of the lipid bilayer to prepare MLs could offer efficient MRI contrast agents, even at very low IO NP concentrations.
Collapse
Affiliation(s)
- Nina Kostevšek
- Department for Nanostructured Materials, Jožef Stefan Institute, 1000 Ljubljana, Slovenia
| | | | - Igor Serša
- Condensed Matter Physics Department, Jožef Stefan Institute, 1000 Ljubljana, Slovenia;
| | - Mateja Erdani Kreft
- Faculty of Medicine, Institute of Cell Biology, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Ilaria Monaco
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, 40136 Bologna, Italy; (I.M.); (M.C.F.)
| | - Mauro Comes Franchini
- Department of Industrial Chemistry “Toso Montanari”, University of Bologna, 40136 Bologna, Italy; (I.M.); (M.C.F.)
| | - Janja Vidmar
- Department for Environmental Sciences, Jožef Stefan Institute, 1000 Ljubljana, Slovenia;
| | - Wafa T. Al-Jamal
- School of Pharmacy, Queen’s University Belfast, Belfast BT9 7BL, UK;
| |
Collapse
|
30
|
Comprehensive Effects of Near-Infrared Multifunctional Liposomes on Cancer Cells. Molecules 2020; 25:molecules25051098. [PMID: 32121482 PMCID: PMC7179136 DOI: 10.3390/molecules25051098] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Multifunctional theranostic systems are a recent important development of medical research. We combined the characteristics of near-infrared luminescent quantum dots and thermosensitive magnetoliposomes to develop a multifunctional nano-diagnostic material. This system is based on near-infrared magnetic thermosensitive liposomes, which encapsulate drugs and can control drug localization and release. After incubating cancer cells with the liposomes, the state of the cells was analyzed in real time by near-infrared imaging. Cell viability was significantly inhibited by heat treatment or alternating magnetic field treatment, which thus improved the anti-cancer properties of the liposomes. In the future, by combining near-infrared imaging technology and an external high-frequency alternating magnetic field, we could not only detect cancer cells noninvasively but also conduct image-guided treatments for cancer.
Collapse
|
31
|
A Review on the Optimal Design of Magnetic Nanoparticle-Based T2 MRI Contrast Agents. MAGNETOCHEMISTRY 2020. [DOI: 10.3390/magnetochemistry6010011] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Relaxivity r2 and thus the contrast efficacy of superparamagnetic nanoparticles (NPs) can be enhanced via either NP’s magnetic properties or coating optimization. Numerous reports can be found about the investigation of the optimal iron oxide nanoparticles (IO NPs) size, shape, crystallinity and composition that yield high saturation magnetization (ms) values and, consequently, high r2 values. Although the use of an appropriate coating can boost up the NPs MRI contrast agent efficiency, this topic has been largely understudied. Therefore, in this review, the factors affording r2 enhancement of spherical magnetic NPs are discussed. Based on the literature, the requirements for an optimal surface coating that may increase r2 values and ensure stability and biocompatibility of NPs are listed. One of the best candidates that fulfil these requirements are liposomes with embedded magnetic NPs, so-called magneto-liposomes. The analysis of the literature elucidated the most appropriate phospholipid compositions for the relaxivity enhancement and for magneto-liposomes in vivo stability. Finally, the future directions in the development of NP-based contrast agents are given. For example, most of the synthetic NPs are recognized and eliminated as a foreign substance by the immune system. To overcome this issue, a design of a biomimetic, cell-membrane-based nanocarrier for contrast agents is proposed. Disguised with cell membranes, NPs or other active components can act as autogenous cells and thus ensure the inherent biocompatibility.
Collapse
|
32
|
Karimi Alavijeh R, Akhbari K. Biocompatible MIL-101(Fe) as a Smart Carrier with High Loading Potential and Sustained Release of Curcumin. Inorg Chem 2020; 59:3570-3578. [PMID: 32091212 DOI: 10.1021/acs.inorgchem.9b02756] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The purpose of this study was the investigation of the potential of MIL-101(Fe) for load and sustained release of curcumin (CCM), as an anticancer drug, with pH stimulus. The reasons for choosing this type of metal-organic framework (MOF) are its high surface area, acceptable stability in a water medium, and its biocompatible components (iron and terephthalic acid) with low toxicity to normal cells. The obtained results from UV-vis analysis confirmed that this MOF is a smart carrier with a higher release rate in acidic pH (pH 5), which is a condition similar to that in cancer cells, than that at pH 7.4 (in normal cells). Therefore, this MOF is a pH-stimulus-controlled release carrier with 56.3% drug loading content and sustained drug release over 22 days. In order to evaluate the cell viability after treatment with free CCM, MIL-101(Fe), and MIL-101(Fe)@CCM, the cytotoxicity investigation using MTT assays was performed against HeLa and HEK 293 cell lines up to 48 h. Obtained results showed that MIL-101(Fe)@CCM exhibited more cell growth inhibition effect on HeLa cells in comparison with HEK 293. One of the reasons for the high loading and sustained release of CCM was surface adsorption of this drug and its interactions with open metal sites in MIL-101(Fe). In the end, the kinetic models of drug release were evaluated, and the obtained results showed that in this case diffusion is the main driving force for the drug release process.
Collapse
Affiliation(s)
| | - Kamran Akhbari
- School of Chemistry, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
33
|
Yang M, Yang W, Chen L, Ding M, Li C, Shi D. A Novel Synthesis of Fe 3O 4@SiO 2@Au@Porous SiO 2 Structure for NIR Irradiation-Induced DOX Release and Cancer Treatment. Dose Response 2020; 18:1559325820906662. [PMID: 32110171 PMCID: PMC7026820 DOI: 10.1177/1559325820906662] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/02/2020] [Accepted: 01/16/2020] [Indexed: 01/29/2023] Open
Abstract
Doxorubicin (DOX) alone or in combination has been widely used for numerous cancers, including breast, lung, bladder, and so on. In this article, a core/shell/shell structured Fe3O4@SiO2@Au@porous SiO2 particles for the drug delivery and release of DOX was demonstrated, with the aid of near-infrared irradiation. Fe3O4 was used to direct the transportation and delivery of the drug-loaded composite to the target tissues and organs under an external magnetic field, the first layer of SiO2 was used for Au nanoparticle attachment, Au acted as the agent for light-thermal conversion, and the porous SiO2 was used to load DOX. The morphology of the nanoparticles was studied by transmission electron microscopy, and the porous structure was characterized by N2 adsorption/desorption curves. The drug delivery system displayed high drug loading capacity, and the release behavior was largely impacted by the environmental pH. Furthermore, the cytotoxicity of Fe3O4@SiO2@Au@porous SiO2 and DOX loaded Fe3O4@SiO2@Au@porous SiO2 was studied through in vitro 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide cell viability assay.
Collapse
Affiliation(s)
- Meng Yang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Wenhua Yang
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Liang Chen
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Mingjian Ding
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Chenhao Li
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, Cangzhou, China
| | - Dongliang Shi
- The Second Department of Thyroid and Breast Surgery, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
34
|
Pan A, Jakaria MG, Meenach SA, Bothun GD. Radiofrequency and Near-Infrared Responsive Core–Shell Nanostructures Using Layersome Templates for Cancer Treatment. ACS APPLIED BIO MATERIALS 2019; 3:273-281. [DOI: 10.1021/acsabm.9b00797] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
35
|
Shen S, Chen Y, Qi S, Zhang X, Qiao C, Wu L. Erythrocyte Membrane Coated Fe3O4 Nanoparticles for Near Infrared Light Responsive Drug Delivery. CHEM LETT 2019. [DOI: 10.1246/cl.190538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Song Shen
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, P. R. China
| | - Ying Chen
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P. R. China
| | - Shunyao Qi
- China Pharmaceutical University, Nanjing 210000, P. R. China
| | - Xin Zhang
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P. R. China
| | - Chen Qiao
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P. R. China
| | - Lin Wu
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, P. R. China
| |
Collapse
|