1
|
Geraldes CFGC. Manganese Oxide Nanoparticles for MRI-Based Multimodal Imaging and Theranostics. Molecules 2024; 29:5591. [PMID: 39683750 PMCID: PMC11643175 DOI: 10.3390/molecules29235591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Manganese-based MRI contrast agents have recently attracted much attention as an alternative to Gd-based compounds. Various nanostructures have been proposed for potential applications in in vivo diagnostics and theranostics. This review is focused on the discussion of different types of Mn oxide-based nanoparticles (MnxOy NPs) obtained at the +2, +3 and +4 oxidation states for MRI, multimodal imaging or theranostic applications. These NPs show favorable magnetic properties, good biocompatibility, and an improved toxicity profile relative to Gd(III)-based nanosystems, showing that the Mn paramagnetic ions offer advantages for the next generation of nanoscale MRI and theranostic contrast agents. Their potential for enhancing relaxivity and MRI contrast effects is illustrated through discussion of selected examples published in the past decade.
Collapse
Affiliation(s)
- Carlos F. G. C. Geraldes
- Department of Life Sciences and Coimbra Chemistry Center-Institute of Molecular Sciences (CQC-IMS), Faculty of Science and Technology, University of Coimbra, 3004-531 Coimbra, Portugal; ; Tel.: +351-967661211
- CIBIT—Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, 3004-531 Coimbra, Portugal
| |
Collapse
|
2
|
Hajebi S, Chamanara M, Nasiri SS, Ghasri M, Mouraki A, Heidari R, Nourmohammadi A. Advances in stimuli-responsive gold nanorods for drug-delivery and targeted therapy systems. Biomed Pharmacother 2024; 180:117493. [PMID: 39353321 DOI: 10.1016/j.biopha.2024.117493] [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: 06/15/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
In recent years, the use of gold nanorods (AuNRs) has garnered considerable attention in biomedical applications due to their unique optical and physicochemical properties. They have been considered as potential tools for the advanced treatment of diseases by various stimuli such as magnetic fields, pH, temperature and light in the fields of targeted therapy, imaging and drug delivery. Their biocompatibility and tunable plasmonic properties make them a versatile platform for a range of biomedical applications. While endogenous stimuli have limited cargo delivery control at specific sites, exogenous stimuli are a more favored approach despite their circumscribed penetration depth for releasing the cargo at the specific target. Dual/multi-stimuli responsive AuNTs can be triggered by multiple stimuli for enhanced control and specificity in biomedical applications. This review provides to provide a summary of the biomedical applications of stimuli-responsive AuNRs, including their endogenous and exogenous properties, as well as their dual/multi-functionality and potential for clinical delivery. This review provides a comprehensive review on the improvement of therapeutic efficacy and the effective control of drug release with AuNRs, highlights AuNRs design strategies in recent years, discusses the advantages or challenges so far in the field of AuNRs. Finally, we have addressed the clinical translation bio-integrated nanoassemblies (CTBNs) in this field.
Collapse
Affiliation(s)
- Sakineh Hajebi
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran; Biomaterial and Medicinal Chemistry Research Center, AJA University of Medical Science, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran; Biomaterial and Medicinal Chemistry Research Center, AJA University of Medical Science, Tehran, Iran
| | - Shadi Sadat Nasiri
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology, Tehran, Iran
| | - Mahsa Ghasri
- Adhesive and Resin Department, Polymer Processing Faculty, Iran Polymer and Petrochemical Institute (IPPI), Tehran, Iran
| | - Alireza Mouraki
- Department of Surface Coating and Corrosion, Institute for Color Science and Technology, Tehran, Iran
| | - Reza Heidari
- Cancer Epidemiology Research Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran; Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran; Biomaterial and Medicinal Chemistry Research Center, AJA University of Medical Science, Tehran, Iran.
| | - Abbas Nourmohammadi
- Clinical Biomechanics and Ergonomics Research Center, AJA University of Medical Sciences, Tehran, Iran; Research Center of Aerospace Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Ju Q, Huang R, Hu R, Fan J, Zhang D, Ding J, Li R. Phytic acid-modified manganese dioxide nanoparticles oligomer for magnetic resonance imaging and targeting therapy of osteosarcoma. Drug Deliv 2023; 30:2181743. [PMID: 36855959 PMCID: PMC9980014 DOI: 10.1080/10717544.2023.2181743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
Osteosarcoma is the most common malignant tumor in the skeletal system with high mortality. Phytic acid (PA) is a natural compound extracted from plant seeds, which shows certain antitumor activity and good bone targeting ability. To develop a novel theranostics for magnetic resonance imaging (MRI) and targeting therapy of osteosarcoma, we employed PA to modify manganese dioxide nanoparticles (MnO2@PA NPs) for osteosarcoma treatment. The MnO2 NPs oligomer was formed by PA modification with uniformed size distribution and negative zeta potential. Fourier-transform infrared spectroscopy, X-ray diffraction, energy dispersive spectroscopy, X-ray photoelectron spectroscopy, and thermogravimetric analysis demonstrated that PA has been successfully modified on MnO2 NPs, and the structure of MnO2@PA NPs is amorphous. In vitro experiments demonstrated that MnO2@PA NPs oligomer can be efficiently internalized by tumor cell, and the internalized NPs can react with H2O2 under acid microenvironment to produce Mn2+ and O2. In vivo experiments demonstrated that MnO2@PA NPs oligomer can passively accumulate in tumor tissue, and the accumulated NPs can produce Mn2+ and O2 for MRI and targeting therapy of osteosarcoma. In conclusion, we prepared a novel bone-targeting nano theranostics for MRI and therapy of osteosarcoma.
Collapse
Affiliation(s)
- Qian Ju
- College of Chemistry, Chongqing Normal University, Chongqing, China,Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Rong Huang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ruimin Hu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junjie Fan
- Department of Clinical Laboratory, the 958th Hospital of Chinese People’s Liberation Army, Chongqing, China
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China,Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China,Dinglin Zhang or Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing400038, China
| | - Jun Ding
- Department of Ultrasonics, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China,Jun Ding Department of Ultrasound, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing400038, China
| | - Rong Li
- College of Chemistry, Chongqing Normal University, Chongqing, China,CONTACT Rong Li College of Chemistry, Chongqing Normal University, Chongqing401331, China
| |
Collapse
|
4
|
Jiang S, Deng X, Luo M, Zhou L, Chai J, Tian C, Yan Y, Luo Z. Pan-cancer analysis identified OAS1 as a potential prognostic biomarker for multiple tumor types. Front Oncol 2023; 13:1207081. [PMID: 37746262 PMCID: PMC10511872 DOI: 10.3389/fonc.2023.1207081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Background 2',5'-oligoadenylate synthetase 1 (OAS1), has been reported as a tumor driver gene in breast carcinoma and pancreatic carcinoma. However, the role of OAS1 in most tumors has not been reported. Methods The original data of 35 tumor types were down load from the TCGA (The Cancer Genome Atlas) database and Human Protein Atlas (HPA) database. TIMER2, Kmplot, UALCAN, and TISIDB tools were used to investigate the expression and function of OAS1, and the role of OAS1 in prognosis, diagnostic value, and immune characteristics of pan-cancer. LUAD and PRAD cell lines, A549, H1975, PC-3 and C4-2 were utilized to perform cell function tests. Results OAS1 expression was up-regulated in 12 tumor types and down-regulated in 2 tumor types. High OAS1 expression was correlated with poor prognosis in 6 tumor types, while high OAS1 expression was correlated with good prognosis in 2 tumor types. OAS1 was correlated with molecular subtypes in 8 tumor types and immune subtypes in 12 tumor types. OAS1 was positively associated with the expression of numerous immune checkpoint genes and tumor mutational burden (TMB). OAS1 had potential diagnostic value in 15 tumor types. Silence of OAS1 significantly inhibited the cell proliferation ability, and promoted G2/M cell cycle arrest of LUAD and PRAD cells. Meanwhile, silence of OAS1 enhanced cisplatin-induced apoptosis of LUAD and PRAD cells, but weakened cell migration. Conclusion This pan-cancer study suggests that OAS1can be used as a molecular biomarker for prognosis in pan-cancer and may play an important role in tumor immune response.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xinzhou Deng
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
- Hubei Key Laboratory of Stem Cell Research, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Ming Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Le Zhou
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jingjing Chai
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Chao Tian
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yutao Yan
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhiguo Luo
- Department of Clinical Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
5
|
Ghadimi Darsajini A, Soleimani M, Mirjani R. The Combination of Photothermal Therapy and Chemotherapy using Alginate-Modified Iron Oxide-Gold Nanohybrids Carrying Cisplatin. J Biomed Phys Eng 2023; 13:117-124. [PMID: 37082548 PMCID: PMC10111104 DOI: 10.31661/jbpe.v0i0.2003-1083] [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: 03/13/2020] [Accepted: 04/20/2020] [Indexed: 04/22/2023]
Abstract
Background Chemotherapy is typically the first-line treatment for the advanced stage of cancers. However, there are shortcomings with respect to conventional chemotherapy that limit therapeutic efficiency, including lack of tumor selectivity, systemic toxicity and drug resistance. Objective A multifunctional nanoplatform was build using of hydrogel co-loaded containing cisplatin and Iron oxide-gold core-shell nanoparticles. The Au shell comprises the light response and the iron core can be utilized as a negative contrast agent in nanocomplex. Material and Methods In this experimental study, KB cells derived from the epithelial cells located in the nasopharynx were exposed to different levels of concentration of hydrogel co-loaded with cisplatin and Iron oxide-gold core-shell nanoparticles. Afterwards, the cytotoxicity was determined using MTT assay. Results The cytotoxicity results showed that this nanoplatforms has potent to create higher cytotoxicity in KB cells than free cisplatin, so that Fe-Au@Alg and Fe-Au@Alg with cisplatin mixed with laser irradiation exhibited a significant reduction in cell viability after 5 min. Conclusion Hydrogel co-loaded with cisplatin and Iron oxide-gold core-shell nanoparticles are stable construct to combine chemo-photothermal therapy. Therefore, they can be used as a computed tomography-traceable nanocarrie, enabling us to monitor the delivery of therapeutics.
Collapse
Affiliation(s)
- Ali Ghadimi Darsajini
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Soleimani
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Ruhollah Mirjani
- Department of Genetics and Advanced Technologies, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Haemmerich D, Ramajayam KK, Newton DA. Review of the Delivery Kinetics of Thermosensitive Liposomes. Cancers (Basel) 2023; 15:cancers15020398. [PMID: 36672347 PMCID: PMC9856714 DOI: 10.3390/cancers15020398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023] Open
Abstract
Thermosensitive liposomes (TSL) are triggered nanoparticles that release the encapsulated drug in response to hyperthermia. Combined with localized hyperthermia, TSL enabled loco-regional drug delivery to tumors with reduced systemic toxicities. More recent TSL formulations are based on intravascular triggered release, where drug release occurs within the microvasculature. Thus, this delivery strategy does not require enhanced permeability and retention (EPR). Compared to traditional nanoparticle drug delivery systems based on EPR with passive or active tumor targeting (typically <5%ID/g tumor), TSL can achieve superior tumor drug uptake (>10%ID/g tumor). Numerous TSL formulations have been combined with various drugs and hyperthermia devices in preclinical and clinical studies over the last four decades. Here, we review how the properties of TSL dictate delivery and discuss the advantages of rapid drug release from TSL. We show the benefits of selecting a drug with rapid extraction by tissue, and with quick cellular uptake. Furthermore, the optimal characteristics of hyperthermia devices are reviewed, and impact of tumor biology and cancer cell characteristics are discussed. Thus, this review provides guidelines on how to improve drug delivery with TSL by optimizing the combination of TSL, drug, and hyperthermia method. Many of the concepts discussed are applicable to a variety of other triggered drug delivery systems.
Collapse
Affiliation(s)
- Dieter Haemmerich
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
- Correspondence:
| | - Krishna K. Ramajayam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Danforth A. Newton
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
7
|
Nanoparticles Design for Theranostic Approach in Cancer Disease. Cancers (Basel) 2022; 14:cancers14194654. [PMID: 36230578 PMCID: PMC9564040 DOI: 10.3390/cancers14194654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Presently, there are no conclusive treatments for many types of cancer, mainly due to the advanced phase of the disease at the time of diagnosis and to the side effects of existing therapies. Present diagnostic and therapeutic procedures need to be improved to supply early detection abilities and perform a more specific therapy with reduced systemic toxicity. In this review, improvements in nanotechnology allowing the design of multifunctional nanoparticles for cancer detection, therapy, and monitoring are reported. Nanoparticles, thanks to the nanomaterials they are made of, can be used as contrast agents for various diagnostic techniques such as MRI, optical imaging, and photoacoustic imaging. Furthermore, when used as drug carriers, they can accumulate in tumor tissues through the passive or/and active targeting, protect encapsulated drugs from degradation, raise tumor exposure to chemotherapeutic agents improving treatment effects. In addition, nanocarriers can simultaneously deliver more than one therapeutic agent enhancing the effectiveness of therapy and can co-deliver imaging and therapy agents to provide integration of diagnostics, therapy, and follow-up. Furthermore, the use of nanocarriers allows to use different therapeutic approaches, such as chemotherapy and hyperthermia to exploit synergistic effects. Theranostic approach to diagnose and treat cancer show a great potential to improve human health, however, despite technological advances in this field, the transfer into clinical practice is still a long way off.
Collapse
|
8
|
PLGA-Based Composites for Various Biomedical Applications. Int J Mol Sci 2022; 23:ijms23042034. [PMID: 35216149 PMCID: PMC8876940 DOI: 10.3390/ijms23042034] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Polymeric materials have been extensively explored in the field of nanomedicine; within them, poly lactic-co-glycolic acid (PLGA) holds a prominent position in micro- and nanotechnology due to its biocompatibility and controllable biodegradability. In this review we focus on the combination of PLGA with different inorganic nanomaterials in the form of nanocomposites to overcome the polymer’s limitations and extend its field of applications. We discuss their physicochemical properties and a variety of well-established synthesis methods for the preparation of different PLGA-based materials. Recent progress in the design and biomedical applications of PLGA-based materials are thoroughly discussed to provide a framework for future research.
Collapse
|
9
|
Yim W, Borum RM, Zhou J, Mantri Y, Wu Z, Zhou J, Jin Z, Creyer M, Jokerst JV. Ultrasmall gold nanorod-polydopamine hybrids for enhanced photoacoustic imaging and photothermal therapy in second near-infrared window. Nanotheranostics 2022; 6:79-90. [PMID: 34976582 PMCID: PMC8671965 DOI: 10.7150/ntno.63634] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 06/19/2021] [Indexed: 12/13/2022] Open
Abstract
Gold nanorods (GNRs) have attracted great interest for photo-mediated biomedicines due to their tunable and high optical absorption, high photothermal conversion efficiency and facile surface modifiability. GNRs that have efficient absorption in second near-infrared (NIR-II) window hold further promise in bio-applications due to low background signal from tissue and deep tissue penetration. However, bare GNRs readily undergo shape deformation (termed as 'melting effect') during the laser illumination losing their unique localized surface plasmon resonance (LSPR) properties, which subsequently leads to PA signal attenuation and decreased photothermal efficiency. Polydopamine (PDA) is a robust synthetic melanin that has broad absorption and high photothermal conversion. Herein, we coated GNRs with PDA to prepare photothermally robust GNR@PDA hybrids for enhanced photo-mediated theranostic agents. Ultrasmall GNRs (SGNRs) and conventional large GNRs (LGNRs) that possess similar LSPR characteristics as well as GNR@PDA hybrids were compared side-by-side in terms of the size-dependent photoacoustic (PA) imaging, photothermal therapy (PTT), and structural stability. In vitro experiments further demonstrated that SGNR@PDA showed 95% ablation of SKOV3 ovarian cancer cells, which is significantly higher than that of LGNRs (66%) and SGNRs (74%). Collectively, our PDA coating strategy represents a rational design for enhanced PA imaging and efficient PTT via a nanoparticle, i.e., nanotheranostics.
Collapse
Affiliation(s)
- Wonjun Yim
- Materials Science and Engineering Program, University of California San Diego, La Jolla, California, 92093, United States
| | - Raina M. Borum
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Jiajing Zhou
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Yash Mantri
- Department of Bioengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Zhuohong Wu
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Jingcheng Zhou
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Zhicheng Jin
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Matthew Creyer
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Jesse V. Jokerst
- Materials Science and Engineering Program, University of California San Diego, La Jolla, California, 92093, United States
- Department of Nanoengineering, University of California San Diego, La Jolla, California, 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California, 92093, United States
| |
Collapse
|
10
|
Liu X, Rong P. Recent Advances of Manganese-Based Hybrid Nanomaterials for Cancer Precision Medicine. Front Oncol 2021; 11:707618. [PMID: 34722253 PMCID: PMC8548572 DOI: 10.3389/fonc.2021.707618] [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: 07/01/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022] Open
Abstract
Cancer precision medicine (CPM) could tailor the best treatment for individual cancer patients, while imaging techniques play important roles in its application. With the characteristics of noninvasion, nonionized, radiation-free, multidimensional imaging function, and real-time monitoring, magnetic resonance imaging (MRI) is an effective way for early tumor detection, and it has become a tower of strength in CPM imaging techniques. Due to linkage with nephrogenic systemic fibrosis (NSF), gadolinium (Gd)-based contrast agent (CA), which was long used in MRI, has been restricted by the Food and Drug Administration (FDA). In this review, we would like to introduce the manganese (Mn)-based CAs that could significantly increase the safety of MRI CAs by realizing more superior performance and functions simultaneously in the diagnosis and treatment of tumors. Also, recent advances in Mn-based hybrid nanomaterials for CPM are summarized and discussed.
Collapse
Affiliation(s)
- Xiaoman Liu
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, China.,Postdoctoral Research Station of Clinical Medicine, Third Xiangya Hospital, Central South University, Changsha, China.,College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengfei Rong
- Department of Radiology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Xi J, Huang Y, Chen J, Zhang J, Gao L, Fan L, Qian X. Artesunate-loaded poly (lactic-co-glycolic acid)/polydopamine-manganese oxides nanoparticles as an oxidase mimic for tumor chemo-catalytic therapy. Int J Biol Macromol 2021; 181:72-81. [PMID: 33771546 DOI: 10.1016/j.ijbiomac.2021.03.124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 01/16/2023]
Abstract
Conventional tumor chemotherapy is limited by its low therapeutic efficacy and side effects, which severely hold back its further application as a first-line agent in clinic. To improve the cure efficacy of cancer, nanozyme with enzyme-like activity has now been extensively investigated as a new strategy for tumor treatment. Herein, an anti-tumor platform based on manganese oxides (MnOx) modified poly (lactic-co-glycolic acid) (PLGA)@polydopamine (PDA) nanoparticles (PP-MnOx NPs) as an oxidase mimic was developed. PP-MnOx NPs could not only produce abundant reactive oxygen species to inhibit tumor growth taking advantage of their oxidase-like activity, but also encapsulate and release antitumor drug (artesunate) to function as chemotherapy, achieving remarkable synergistic chemo-catalytic therapeutic effects. As an oxidase mimics, PP-MnOx NPs induced the decrease of mitochondrial membrane potential, down-regulation of Bcl-2, as well as activation of Bax and Caspase-3, demonstrating that the apoptosis triggered by PP-MnOx NPs was mediated via mitochondrial pathways. Importantly, the artesunate in PP-MnOx NPs further promoted this apoptosis. In addition, Mn ions released from PP-MnOx NPs facilitated the tumor-microenvironment-specific T1-weighted magnetic resonance imaging. Taken together, this study well clarifies the antitumor mechanism of artesunate-loaded PP-MnOx NPs and offer a synergistic chemo-catalytic strategy for tumor theranostics.
Collapse
Affiliation(s)
- Juqun Xi
- Institute of Translational Medicine, Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, Jiangsu 225009, China
| | - Yaling Huang
- Institute of Translational Medicine, Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jie Chen
- Institute of Translational Medicine, Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Jingjing Zhang
- Institute of Translational Medicine, Department of Pharmacology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
| | - Lizeng Gao
- CAS Engineering Laboratory for Nanozyme, Institute of Biophysis, Chinese Academy of Sciences, Beijing 100101, China
| | - Lei Fan
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, Jiangsu 225002, China.
| | - Xiaodong Qian
- Department of Cardiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
12
|
Liao S, Yue W, Cai S, Tang Q, Lu W, Huang L, Qi T, Liao J. Improvement of Gold Nanorods in Photothermal Therapy: Recent Progress and Perspective. Front Pharmacol 2021; 12:664123. [PMID: 33967809 PMCID: PMC8100678 DOI: 10.3389/fphar.2021.664123] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/24/2021] [Indexed: 02/05/2023] Open
Abstract
Cancer is a life-threatening disease, and there is a significant need for novel technologies to treat cancer with an effective outcome and low toxicity. Photothermal therapy (PTT) is a noninvasive therapeutic tool that transports nanomaterials into tumors, absorbing light energy and converting it into heat, thus killing tumor cells. Gold nanorods (GNRs) have attracted widespread attention in recent years due to their unique optical and electronic properties and potential applications in biological imaging, molecular detection, and drug delivery, especially in the PTT of cancer and other diseases. This review summarizes the recent progress in the synthesis methods and surface functionalization of GNRs for PTT. The current major synthetic methods of GNRs and recently improved measures to reduce toxicity, increase yield, and control particle size and shape are first introduced, followed by various surface functionalization approaches to construct a controlled drug release system, increase cell uptake, and improve pharmacokinetics and tumor-targeting effect, thus enhancing the photothermal effect of killing the tumor. Finally, a brief outlook for the future development of GNRs modification and functionalization in PTT is proposed.
Collapse
Affiliation(s)
- Shengnan Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wang Yue
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuning Cai
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Weitong Lu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lingxiao Huang
- Department of Radiation Biology, Radiation Oncology Key Laboratory of Sichuan Province, Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Tingting Qi
- Department of Radiation Biology, Radiation Oncology Key Laboratory of Sichuan Province, Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
El-Sherbiny RH, Hassan MM, El-Hossary WH, Shata MS, Darwish WM. Folate-targeted polymeric nanoparticles for efficient dual (chemo-photothermal) therapy of oral squamous carcinoma. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2020.1725756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Randa H. El-Sherbiny
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Magda M. Hassan
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Wafaa H. El-Hossary
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Mona S. Shata
- Department of Oral Pathology, Faculty of Dentistry, Suez Canal University, Ismailia, Egypt
| | - Wael M. Darwish
- Department of Polymers and Pigments, National Research Centre, Giza, Dokki, Egypt
| |
Collapse
|
14
|
Shi X, Perry HL, Wilton-Ely JDET. Strategies for the functionalisation of gold nanorods to reduce toxicity and aid clinical translation. Nanotheranostics 2021; 5:155-165. [PMID: 33564615 PMCID: PMC7868005 DOI: 10.7150/ntno.56432] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022] Open
Abstract
Gold nanorods (GNRs) show great promise as photothermal therapy agents due to their remarkable ability to convert light into heat. In most cases, gold nanorods are synthesised via a seed-mediated method assisted by surfactants. However, the toxicity of these surfactants, principally cetrimonium ions, has prevented GNRs from being used more widely in vivo. To address this issue, various detoxification and functionalisation approaches have been proposed in recent years to replace or cover surfactant coatings on the gold surface. In this short review, the advantages and limitations of each approach are examined in the context of the recent progress made towards the design of GNRs suitable for use in the body.
Collapse
Affiliation(s)
- Xin Shi
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, London W12 0BZ, United Kingdom
| | - Hannah L Perry
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, London W12 0BZ, United Kingdom
| | - James D E T Wilton-Ely
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, London W12 0BZ, United Kingdom
| |
Collapse
|
15
|
Wang N, Fuh JYH, Dheen ST, Senthil Kumar A. Synthesis methods of functionalized nanoparticles: a review. Biodes Manuf 2021. [DOI: 10.1007/s42242-020-00106-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Darwish W. Polymers for enhanced photodynamic cancer therapy: Phthalocyanines as a photosensitzer model. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.5154] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Wael Darwish
- Laser Technology Group, Center of Excellence for Advanced Sciences; Department of Polymers and Pigments National Research Centre Giza Egypt
| |
Collapse
|
17
|
Soratijahromi E, Mohammadi S, Dehdari Vais R, Azarpira N, Sattarahmady N. Photothermal/sonodynamic therapy of melanoma tumor by a gold/manganese dioxide nanocomposite: In vitro and in vivo studies. Photodiagnosis Photodyn Ther 2020; 31:101846. [DOI: 10.1016/j.pdpdt.2020.101846] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/18/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
|
18
|
Cheng YH, He C, Riviere JE, Monteiro-Riviere NA, Lin Z. Meta-Analysis of Nanoparticle Delivery to Tumors Using a Physiologically Based Pharmacokinetic Modeling and Simulation Approach. ACS NANO 2020; 14:3075-3095. [PMID: 32078303 PMCID: PMC7098057 DOI: 10.1021/acsnano.9b08142] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/20/2020] [Indexed: 05/18/2023]
Abstract
Numerous studies have engineered nanoparticles with different physicochemical properties to enhance the delivery efficiency to solid tumors, yet the mean and median delivery efficiencies are only 1.48% and 0.70% of the injected dose (%ID), respectively, according to a study using a nonphysiologically based modeling approach based on published data from 2005 to 2015. In this study, we used physiologically based pharmacokinetic (PBPK) models to analyze 376 data sets covering a wide range of nanomedicines published from 2005 to 2018 and found mean and median delivery efficiencies at the last sampling time point of 2.23% and 0.76%ID, respectively. Also, the mean and median delivery efficiencies were 2.24% and 0.76%ID at 24 h and were decreased to 1.23% and 0.35%ID at 168 h, respectively, after intravenous administration. While these delivery efficiencies appear to be higher than previous findings, they are still quite low and represent a critical barrier in the clinical translation of nanomedicines. We explored the potential causes of this poor delivery efficiency using the more mechanistic PBPK perspective applied to a subset of gold nanoparticles and found that low delivery efficiency was associated with low distribution and permeability coefficients at the tumor site (P < 0.01). We also demonstrate how PBPK modeling and simulation can be used as an effective tool to investigate tumor delivery efficiency of nanomedicines.
Collapse
Affiliation(s)
- Yi-Hsien Cheng
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Chunla He
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Jim E. Riviere
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- 1Data
Consortium, Kansas State University, Manhattan, Kansas 66506, United States
| | - Nancy A. Monteiro-Riviere
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| | - Zhoumeng Lin
- Institute
of Computational Comparative Medicine (ICCM), Department of Anatomy
and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
- Nanotechnology
Innovation Center of Kansas State (NICKS), Department of Anatomy and
Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas 66506, United States
| |
Collapse
|
19
|
Darwish WMA, Bayoumi NA. Gold nanorod-loaded (PLGA-PEG) nanocapsules as near-infrared controlled release model of anticancer therapeutics. Lasers Med Sci 2020; 35:1729-1740. [PMID: 31965353 DOI: 10.1007/s10103-020-02964-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/10/2020] [Indexed: 12/17/2022]
Abstract
Despite of high in vitro anticancer efficacy of many chemotherapeutics, their in vivo use is limited due to lack of biocompatibility and tumor targeting. Near-infrared (NIR) photothermally induced phase transition of PLGA-PEG regime was utilized for developing highly efficient photoresponsive drug delivery systems. Co-encapsulation of plasmonic gold nanorods (GNRs), as NIR-trigger, with the novel and highly efficient anticancer drug N'-(2-Methoxybenzylidene)-3-methyl-1-phenyl-H-Thieno[2,3-c]Pyrazole-5-Carbohyd-razide (MTPC) produced NIR-responsive biodegradable polymeric (PLGA-b-PEG) nanocapsules. This remotely controllable drug release significantly enhanced both biodistribution and pharmacokinetics of the hydrophobic drug. Intravenous (IV) injection of the prepared nanocapsules (MTPC/GNRs@PLGA-PEG) to tumor-bearing mice followed by extracorporeal exposure of the tumor to NIR light resulted in highly selective drug accumulation at the tumor sites. In vivo biodistribution and pharmacokinetics utilizing iodine-131 drug-radiolabelling technique revealed a maximum target to non-target ratio (T/NT) of 5.8, 4 h post-injection with maximum drug level in the tumor (6.3 ± 0.6% of the injected dose). Graphical abstract.
Collapse
Affiliation(s)
- Wael Mahmoud Ahmed Darwish
- Department of Polymers and Pigments, National Research Centre, Elbuhouth Street, Dokki, Giza, 12622, Egypt.
| | - Noha A Bayoumi
- Department of Radiolabeled Compounds, Hot Lab Centre, Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
20
|
Xu W, Lin Q, Yin Y, Xu D, Huang X, Xu B, Wang G. A Review on Cancer Therapy Based on the Photothermal Effect of Gold Nanorod. Curr Pharm Des 2020; 25:4836-4847. [DOI: 10.2174/1381612825666191216150052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 11/29/2019] [Indexed: 02/05/2023]
Abstract
Background:
Cancer causes millions of deaths and huge economic losses every year. The currently
practiced methods for cancer therapy have many defects, such as side effects, low curate rate, and discomfort for
patients.
Objective:
Herein, we summarize the applications of gold nanorods (AuNRs) in cancer therapy based on their
photothermal effect-the conversion of light into local heat under irradiation.
Methods:
The recent advances in the synthesis and regulation of AuNRs, and facile surface functionalization
further facilitate their use in cancer treatment. For cancer therapy, AuNRs need to be modified or coated with
biocompatible molecules (e.g. polyethylene glycol) and materials (e.g. silicon) to reduce the cytotoxicity and
increase their biocompatibility, stability, and retention time in the bloodstream. The accumulation of AuNRs in
cancerous cells and tissues is due to the high leakage in tumors or the specific interaction between the cell surface
and functional molecules on AuNRs such as antibodies, aptamers, and receptors.
Results:
AuNRs are employed not only as therapeutics to ablate tumors solely based on the heat produced under
laser that could denature protein and activate the apoptotic pathway, but also as synergistic therapies combined
with photodynamic therapy, chemotherapy, and gene therapy to kill cancer more efficiently. More importantly,
other materials like TiO2, graphene oxide, and silicon, etc. are incorporated on the AuNR surface for multimodal
cancer treatment with high drug loadings and improved cancer-killing efficiency. To highlight their applications
in cancer treatment, examples of therapeutic effects both in vitro and in vivo are presented.
Conclusion:
AuNRs have potential applications for clinical cancer therapy.
Collapse
Affiliation(s)
- Weizhen Xu
- Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha, 410004, China
| | - Qinlu Lin
- Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha, 410004, China
| | - Yueqin Yin
- Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha, 410004, China
| | - Dong Xu
- Hunan Key Laboratory of Processed Food for Special Medical Purpose, Central South University of Forestry and Technology, Changsha, 410004, China
| | - Xiaohui Huang
- Hunan Edible Fungi Institute, Changsha, 410004, China
| | - Bucheng Xu
- Wangcheng Commodity Inspection Center, Changsha, 410200, China
| | - Guangwei Wang
- Biomedical Research Center, Hunan University of Medicine, Huaihua, 418000, China
| |
Collapse
|
21
|
Nanoengineering of Gold Nanoparticles: Green Synthesis, Characterization, and Applications. CRYSTALS 2019. [DOI: 10.3390/cryst9120612] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The fundamental aspects of the manufacturing of gold nanoparticles (AuNPs) are discussed in this review. In particular, attention is devoted to the development of a simple and versatile method for the preparation of these nanoparticles. Eco-friendly synthetic routes, such as wet chemistry and biosynthesis with the aid of polymers, are of particular interest. Polymers can act as reducing and/or capping agents, or as soft templates leading to hybrid nanomaterials. This methodology allows control of the synthesis and stability of nanomaterials with novel properties. Thus, this review focus on a fundamental study of AuNPs properties and different techniques to characterize them, e.g., Transmission Electron Microscopy (TEM), Atomic Force Microscopy (AFM), UV-Visible spectroscopy, Dynamic Light Scattering (DLS), X-Ray Diffraction (XRD), X-Ray Photoelectron Spectroscopy, Small-angle X-Ray Scattering (SAXS), and rheology. Recently, AuNPs obtained by “green” synthesis have been applied in catalysis, in medicine, and as antibacterials, sensors, among others.
Collapse
|
22
|
Keratin nanoparticles co-delivering Docetaxel and Chlorin e6 promote synergic interaction between chemo- and photo-dynamic therapies. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 199:111598. [PMID: 31465971 DOI: 10.1016/j.jphotobiol.2019.111598] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/23/2019] [Accepted: 08/18/2019] [Indexed: 12/26/2022]
Abstract
The combination of chemotherapy and photodynamic therapy (PDT) is considered a valuable strategy for increasing therapeutic response in cancer treatment, and the re-formulation of pharmaceuticals in biocompatible nanoparticles (NPs) is particularly appealing for the possibility of co-loading drugs exerting cytotoxicity by different mechanisms, with the aim to produce synergic effects. We report the in-water synthesis of a novel keratin-based nanoformulation for the co-delivery of the antimitotic Docetaxel (DTX) and the photosensitizer Chlorin e6 (Ce6). The drug-induced aggregation method allowed the formation of monodisperse NPs (DTX/Ce6-KNPs) with an average diameter of 133 nm and loaded with a drug ratio of 1:1.8 of Ce6 vs DTX. The efficacy of DTX/Ce6-KNPs was investigated in vitro in monolayers and spheroids of DTX-sensitive HeLa (HeLa-P) and DTX-resistant HeLa (HeLa-R) cells. In monolayers, the cytotoxic effects of DTX/Ce6-KNPs toward HeLa-P cells were comparable to those induced by free DTX + Ce6, while in HeLa-R cells the drug co-loading in KNPs produced synergic interaction between chemotherapy and PDT. Moreover, as respect to monotherapies, DTX/Ce6-KNPs induced stronger cytotoxicity to both HeLa-P and HeLa-R multicellular spheroids and reduced their volumes up to 50%. Overall, the results suggest that KNPs are very promising systems for the co-delivery of chemotherapeutics and PSs, favoring synergic interactions between PDT and chemotherapy.
Collapse
|
23
|
Zhang Y, Wang H, Jia X, Du S, Yin Y, Zhang X. Cascade catalytic nanoplatform for enhanced starvation and sonodynamic therapy. J Drug Target 2019; 28:195-203. [PMID: 31282750 DOI: 10.1080/1061186x.2019.1641507] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background: Sonodynamic therapy (SDT) has emerged as an alternative to the traditional treatments of cancer. However, the oxygen consumption induced by SDT and glucose oxidase (GOx) mediated starvation therapy would worsen the hypoxic tumor environment, which further impeded therapeutic efficacy. Purpose: To develop a nanoplatform and investigate its anti-cancer mechanism for enhanced starvation and SDT.Methods: We constructed a cascade catalytic nanoplatform based on GOx modified the mesoporous MnO2 NPs loaded with hematoporphyrin monomethyl ether (HMME), which were designated as GOx-MnO2/HMME. We characterized them for their catalytic activity, and investigate the magnetic resonance imaging and anti-tumor efficiency in vitro and in vivo.Results: MnO2 NPs with catalase-like activity could oxidize H2O2 under acid condition to produce O2, which not only in turn was supplied to the glucose-depletion reaction for an efficient starvation therapy, but also enhanced the 1O2 generation for HMME mediated SDT effect. In addition, the released Mn2+ ions in the system were able to enhance the MRI signal. Both in vitro and in vivo experiments suggested the cascade catalytic-therapeutic effect between GOx, MnO2 NPs and HMME, demonstrating the enhanced starvation and SDT.
Collapse
Affiliation(s)
- Yingjie Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiling Wang
- School of Pharmaceutical Sciences, Zhengzhou Railway Vocational and Technical College, Zhengzhou, China
| | - Xuedong Jia
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuzhang Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanyan Yin
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Xiaojian Zhang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
|
25
|
Alamzadeh Z, Beik J, Pirhajati Mahabadi V, Abbasian Ardekani A, Ghader A, Kamrava SK, Shiralizadeh Dezfuli A, Ghaznavi H, Shakeri-Zadeh A. Ultrastructural and optical characteristics of cancer cells treated by a nanotechnology based chemo-photothermal therapy method. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 192:19-25. [PMID: 30665146 DOI: 10.1016/j.jphotobiol.2019.01.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/21/2018] [Accepted: 01/12/2019] [Indexed: 12/21/2022]
Abstract
The current chemotherapy method demonstrates the need for improvement in terms of efficacy and safety. Given the beneficiary effect of heat in combination with chemotherapy, the purpose of this study is to develop a multifunctional nanoplatform by co-incorporating gold nanoparticles (AuNPs) as photothermal agent and cisplatin as anticancer drug into alginate hydrogel (named as ACA) to enable concurrent thermo-chemotherapy. The in vitro cytotoxicity experiment showed that the as-developed nanocomplex was able to induce greater cytotoxicity in KB human nasopharyngeal cancer cells compared to free cisplatin at the same concentration. Moreover, the interaction of ACA and laser irradiation acted synergistically and resulted in higher cell death rate compared to separate application of photothermal therapy and chemotherapy. The micrograph of KB cells also revealed that ACA was able to selectively accumulate into the mitochondria, so that laser irradiation of KB cells pre-treated with ACA resulted in intensive morphological damages such as plasma membrane disruption, chromatin condensation, autophagic vacuoles formation and organelle degeneration. Moreover, the sign and magnitude of optical nonlinear refractive index measured by Z-scan technique was shown to be significantly altered in cells exposed to ACA with and without laser irradiation. Consequently, the nanocomplex developed herein could be a promising platform to combine photothermal therapy and chemotherapy effectively, thereby achieving synergistic therapeutic outcome.
Collapse
Affiliation(s)
- Zahra Alamzadeh
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Jaber Beik
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Vahid Pirhajati Mahabadi
- Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Abbasian Ardekani
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Ghader
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - S Kamran Kamrava
- ENT and Head & Neck Research Center and Department, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Amin Shiralizadeh Dezfuli
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Habib Ghaznavi
- Zahedan University of Medical Sciences (ZaUMS), Zahedan, Iran.
| | - Ali Shakeri-Zadeh
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran; ENT and Head & Neck Research Center and Department, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
26
|
Ray S, Li Z, Hsu CH, Hwang LP, Lin YC, Chou PT, Lin YY. Dendrimer- and copolymer-based nanoparticles for magnetic resonance cancer theranostics. Theranostics 2018; 8:6322-6349. [PMID: 30613300 PMCID: PMC6299700 DOI: 10.7150/thno.27828] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 09/20/2018] [Indexed: 01/06/2023] Open
Abstract
Cancer theranostics is one of the most important approaches for detecting and treating patients at an early stage. To develop such a technique, accurate detection, specific targeting, and controlled delivery are the key components. Various kinds of nanoparticles have been proposed and demonstrated as potential nanovehicles for cancer theranostics. Among them, polymer-like dendrimers and copolymer-based core-shell nanoparticles could potentially be the best possible choices. At present, magnetic resonance imaging (MRI) is widely used for clinical purposes and is generally considered the most convenient and noninvasive imaging modality. Superparamagnetic iron oxide (SPIO) and gadolinium (Gd)-based dendrimers are the major nanostructures that are currently being investigated as nanovehicles for cancer theranostics using MRI. These structures are capable of specific targeting of tumors as well as controlled drug or gene delivery to tumor sites using pH, temperature, or alternating magnetic field (AMF)-controlled mechanisms. Recently, Gd-based pseudo-porous polymer-dendrimer supramolecular nanoparticles have shown 4-fold higher T1 relaxivity along with highly efficient AMF-guided drug release properties. Core-shell copolymer-based nanovehicles are an equally attractive alternative for designing contrast agents and for delivering anti-cancer drugs. Various copolymer materials could be used as core and shell components to provide biostability, modifiable surface properties, and even adjustable imaging contrast enhancement. Recent advances and challenges in MRI cancer theranostics using dendrimer- and copolymer-based nanovehicles have been summarized in this review article, along with new unpublished research results from our laboratories.
Collapse
Affiliation(s)
- Sayoni Ray
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Zhao Li
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Chao-Hsiung Hsu
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Lian-Pin Hwang
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Ying-Chih Lin
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Pi-Tai Chou
- Department of Chemistry, National Taiwan University, Taipei 10617, Taiwan
| | - Yung-Ya Lin
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
27
|
Wang L, Niu M, Zheng C, Zhao H, Niu X, Li L, Hu Y, Zhang Y, Shi J, Zhang Z. A Core-Shell Nanoplatform for Synergistic Enhanced Sonodynamic Therapy of Hypoxic Tumor via Cascaded Strategy. Adv Healthc Mater 2018; 7:e1800819. [PMID: 30303621 DOI: 10.1002/adhm.201800819] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/13/2018] [Indexed: 01/01/2023]
Abstract
Sonodynamic therapy (SDT) always causes tumor hypoxia aggravation which can induce malignant cell proliferation and drug resistance. To overcome these disadvantages, a cascaded drug delivery system (Lipo/HMME/ACF@MnO2 -AS1411) is constructed for synergistic enhanced sonodynamic therapy. First, hematoporphyrin monomethyl ether (HMME) and acriflavine (ACF) are encapsulated in the lipid layers and the inner aqueous cores of the liposomes, respectively. Then the ultrathin manganese dioxide (MnO2 ) nanosheets are coated on the surface of the liposomes by using KMnO4 and polyethylene glycol through "one step reduction and modification" method. Furthermore, the nanoparticles are decorated with tumor-targeting AS1411 aptamer through the phosphate groups on the DNA strand which can bind to Mn sites to obtain Lipo/HMME/ACF@MnO2 -AS1411 delivery system. Herein, HMME can act as a sonosensitizer, and ACF is used to prevent the formation of HIF-1α/HIF-1β dimerization to overcome the negative effects after SDT. The Lipo/HMME/ACF@MnO2 -AS1411 delivery system has multiple functions, including codelivery of HMME and ACF, pH/glutathione/ultrasound triple responses, synergistic cascaded enhancement of SDT, precise tumor-targeting, and magnetic resonance imaging. The in vitro and in vivo results suggest that the Lipo/HMME/ACF@MnO2 -AS1411 delivery system is a promising core-shell nanoplatform for synergistic enhancement of sonodynamic therapy, which can provide a new approach in the related research fields.
Collapse
Affiliation(s)
- Lei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Mengya Niu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Cuixia Zheng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Hongjuan Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Xiuxiu Niu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Li Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Yujie Hu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Yingjie Zhang
- The First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, P. R. China
| |
Collapse
|
28
|
García-Hevia L, Bañobre-López M, Gallo J. Recent Progress on Manganese-Based Nanostructures as Responsive MRI Contrast Agents. Chemistry 2018; 25:431-441. [PMID: 29999200 DOI: 10.1002/chem.201802851] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/09/2018] [Indexed: 01/10/2023]
Abstract
Manganese-based nanostructured contrast agents (CAs) entered the field of medical diagnosis through magnetic resonance imaging (MRI) some years ago. Although some of these Mn-based CAs behave as classic T1 contrast enhancers in the same way as clinical Gd-based molecules do, a new type of Mn nanomaterials have been developed to improve MRI sensitivity and potentially gather new functional information from tissues by using traditional T1 contrast enhanced MRI. These nanomaterials have been designed to respond to biological environments, mainly to pH and redox potential variations. In many cases, the differences in signal generation in these responsive Mn-based nanostructures come from intrinsic changes in the magnetic properties of Mn cations depending on their oxidation state. In other cases, no changes in the nature of Mn take place, but rather the nanomaterial as a whole responds to the change in the environment through different mechanisms, including changes in integrity and hydration state. This review focusses on the chemistry and MR performance of these responsive Mn-based nanomaterials.
Collapse
Affiliation(s)
- Lorena García-Hevia
- Advanced (Magnetic) Theranostic Nanostructures Laboratory, Department of Life Sciences, INL-International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga, 4715-330, Braga, Portugal
| | - Manuel Bañobre-López
- Advanced (Magnetic) Theranostic Nanostructures Laboratory, Department of Life Sciences, INL-International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga, 4715-330, Braga, Portugal
| | - Juan Gallo
- Advanced (Magnetic) Theranostic Nanostructures Laboratory, Department of Life Sciences, INL-International Iberian Nanotechnology Laboratory, Av. Mestre José Veiga, 4715-330, Braga, Portugal
| |
Collapse
|
29
|
Ghaznavi H, Hosseini-Nami S, Kamrava SK, Irajirad R, Maleki S, Shakeri-Zadeh A, Montazerabadi A. Folic acid conjugated PEG coated gold-iron oxide core-shell nanocomplex as a potential agent for targeted photothermal therapy of cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1594-1604. [PMID: 28994325 DOI: 10.1080/21691401.2017.1384384] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study reports the synthesis and characterization of poly(ethylene glycol) coated gold@iron oxide core-shell nanoparticles conjugated with folic acid (FA-PEG-Au@IONP). Also, targeted therapeutic properties of such a nanocomplex were studied on human nasopharyngeal carcinoma cell line KB and human breast adenocarcinoma cell line MCF-7 in vitro. The synthesized nanocomplex was characterized by transmission electron microscopy (TEM), dynamic light scattering (DLS), UV-Vis spectroscopy, vibrating sample magnetometry (VSM), and Fourier transform infrared (FTIR) spectroscopy. The photothermal effects of nanocomplex on both KB and MCF-7 cell lines were studied. Cell death and apoptosis were evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry using an annexin V-fluorescein isothiocyanate/propidiumiodide apoptosis detection kit. It was found that nanocomplex is spherical in shape and its size is approximately 60 nm. UV-vis spectrum showed that nanocomplex has appropriate absorption near infrared region. FTIR spectra obtained from nanocomplex before and after conjugation with FA confirmed the formation of folate conjugated nanocomplex. Significant cell lethality was observed for KB (∼62%) and MCF-7 (∼33%) cells following photothermal therapy. Also, it was found that majority of the cell deaths were related to apoptosis process. It can be concluded that, the synthesized nanocomplex is an effective and promising multifunctional nanoplatform for targeted photothermal therapy of cancer.
Collapse
Affiliation(s)
- Habib Ghaznavi
- a Health Promotion Research Centre, Zahedan University of Medical Sciences , Zahedan , Iran
| | - Samira Hosseini-Nami
- b Department of Medical Physics , School of Medicine, Iran University of Medical Sciences , Tehran , Iran
| | - S Kamran Kamrava
- c Department of ENT, Head and Neck Research Centre , Iran University of Medical Sciences , Tehran , Iran
| | - Rasoul Irajirad
- d Département de chimie, faculté des sciences et technologies , Université Claude Bernard Lyon 1 , Lyon , France
| | - Shayan Maleki
- c Department of ENT, Head and Neck Research Centre , Iran University of Medical Sciences , Tehran , Iran
| | - Ali Shakeri-Zadeh
- b Department of Medical Physics , School of Medicine, Iran University of Medical Sciences , Tehran , Iran.,e Radiation Biology Research Centre, Iran University of Medical Sciences , Tehran , Iran
| | - Alireza Montazerabadi
- f Department of Medical Physics, School of Medicine , Gonabad University of Medical Sciences , Gonabad, Iran
| |
Collapse
|