1
|
Liu Y, Hinnant B, Chen S, Tao H, Huang Z, Qian M, Zhou M, Han Z, Han ZC, Zhang J, Li Z. Hyaluronic acid-modified extracellular vesicles for targeted doxorubicin delivery in hepatocellular carcinoma. Exp Cell Res 2024; 443:114332. [PMID: 39551341 DOI: 10.1016/j.yexcr.2024.114332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Hepatocellular carcinoma (HCC), a prevalent and deadly cancer, poses a significant challenge with current treatments due to limitations such as poor stability, off-target effects, and severe side effects. Extracellular vesicles (EVs), derived from tumor cells, have the remarkable ability to home back to their cells of origin and can serve as Trojan horses for drug delivery. CD44, a cell surface glycoprotein, promotes cancer stem cell-like properties and is linked to poor prognosis and resistance to chemotherapy in HCC. Therefore, targeting CD44-expressing HCC cells is of interest in the development of novel therapeutic strategies for the treatment of HCC. In this study, we developed tumor cell-derived EVs (TEVs) functionalized with hyaluronic acid (HA) to serve as natural carriers for the precise delivery of doxorubicin (Dox), which specifically targets HCC cells expressing CD44. Our results demonstrated that HA-engineered EVs (HA-EVs) significantly enhanced Dox accumulation within HCC cells. In a mouse model, HA-EVs effectively delivered Dox to tumors, suppressing their growth and progression while minimizing systemic toxicity. This study demonstrates the potential of HA-functionalized EVs as a novel and targeted therapeutic platform for HCC, offering a valuable strategy for improving drug delivery and patient outcomes. This study presents a promising strategy to advance targeted chemotherapy for HCC and address the challenges associated with conventional treatments. Engineered HA-functionalized EVs offer a tailored and efficient approach to increase drug delivery precision, underscoring their potential as a novel therapeutic platform in the realm of HCC treatment.
Collapse
Affiliation(s)
- Yue Liu
- School of Medicine, Nankai University, China; The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China
| | - Benjamin Hinnant
- Division of Biological Sciences, University of California San Diego, USA
| | - Shang Chen
- School of Medicine, Nankai University, China
| | - Hongyan Tao
- School of Medicine, Nankai University, China
| | - Ziyu Huang
- School of Medicine, Nankai University, China
| | - Meng Qian
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China
| | - Manqian Zhou
- Department of Radiation Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Zhibo Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China; Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health Biotech Co., Beijing, China
| | - Zhong-Chao Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China; Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health Biotech Co., Beijing, China
| | - Jun Zhang
- Department of Anesthesiology and Pain Medical Center, Tianjin Union Medical Center, Nankai University, Tianjin, China.
| | - Zongjin Li
- School of Medicine, Nankai University, China; The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China; Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou, China.
| |
Collapse
|
2
|
Sun H, Wang X, Guo Z, Hu Z, Yin Y, Duan S, Jia W, Lu W, Hu J. Fe 3O 4 Nanoparticles That Modulate the Polarisation of Tumor-Associated Macrophages Synergize with Photothermal Therapy and Immunotherapy (PD-1/PD-L1 Inhibitors) to Enhance Anti-Tumor Therapy. Int J Nanomedicine 2024; 19:7185-7200. [PMID: 39050876 PMCID: PMC11268759 DOI: 10.2147/ijn.s459400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/22/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Traditional surgical resection, radiotherapy, and chemotherapy have been the treatment options for patients with head and neck squamous cell carcinoma (HNSCC) over the past few decades. Nevertheless, the five-year survival rate for patients has remained essentially unchanged, and research into treatments has been relatively stagnant. The combined application of photothermal therapy (PTT) and immunotherapy for treating HNSCC has considerable potential. Methods Live-dead cell staining and CCK-8 assays proved that Fe3O4 nanoparticles are biocompatible in vitro. In vitro, cellular experiments utilized flow cytometry and immunofluorescence staining to verify the effect of Fe3O4 nanoparticles on the polarisation of tumor-associated macrophages. In vivo, animal experiments were conducted to assess the inhibitory effect of Fe3O4 nanoparticles on tumor proliferation under the photothermal effect in conjunction with BMS-1. Tumour tissue sections were stained to observe the effects of apoptosis and the inhibition of tumor cell proliferation. The histological damage to animal organs was analyzed by hematoxylin and eosin (H&E) staining. Results The stable photothermal properties of Fe3O4 nanoparticles were validated by in vitro cellular and in vivo animal experiments. Fe3O4 photothermal action not only directly triggered immunogenic cell death (ICD) and enhanced the immunogenicity of the tumor microenvironment but also regulated the expression of tumor-associated macrophages (TAMs), up-regulating CD86 and down-regulating CD206 to inhibit tumor growth. The PD-1/PD-L1 inhibitor promoted tumor suppression, and reduced tumor recurrence and metastasis. In vivo studies demonstrated that the photothermal action exhibited a synergistic effect when combined with immunotherapy, resulting in significant suppression of primary tumors and an extension of survival. Conclusion In this study, we applied Fe3O4 photothermolysis in a biomedical context, combining photothermolysis with immunotherapy, exploring a novel pathway for treating HNSCC and providing a new strategy for effectively treating HNSCC.
Collapse
Affiliation(s)
- Haishui Sun
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People’s Republic of China
| | - Xiao Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of D&A for Metal Functional Materials, School of Materials Science and Engineering, Tongji University, Shanghai, People’s Republic of China
| | - Zhaoyang Guo
- School of Stomatology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Zhenrong Hu
- Department of Stomatology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yuanchen Yin
- School of Stomatology, Weifang Medical University, Weifang, Shandong Province, People’s Republic of China
| | - Shuhan Duan
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, Shanghai Ninth People’s Hospital, College of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Research Institute of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Wenwen Jia
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People’s Republic of China
| | - Wei Lu
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of D&A for Metal Functional Materials, School of Materials Science and Engineering, Tongji University, Shanghai, People’s Republic of China
| | - Jingzhou Hu
- Department of Oral and Maxillofacial - Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, Zhang Zhiyuan Academician Workstation, Hainan Western Central Hospital, Shanghai Ninth People’s Hospital, Danzhou, Hainan, People’s Republic of China
| |
Collapse
|
3
|
Lin P, Lu Y, Zheng J, Lin Y, Zhao X, Cui L. Strategic disruption of cancer's powerhouse: precise nanomedicine targeting of mitochondrial metabolism. J Nanobiotechnology 2024; 22:318. [PMID: 38849914 PMCID: PMC11162068 DOI: 10.1186/s12951-024-02585-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/26/2024] [Indexed: 06/09/2024] Open
Abstract
Mitochondria occupy a central role in the biology of most eukaryotic cells, functioning as the hub of oxidative metabolism where sugars, fats, and amino acids are ultimately oxidized to release energy. This crucial function fuels a variety of cellular activities. Disruption in mitochondrial metabolism is a common feature in many diseases, including cancer, neurodegenerative conditions and cardiovascular diseases. Targeting tumor cell mitochondrial metabolism with multifunctional nanosystems emerges as a promising strategy for enhancing therapeutic efficacy against cancer. This review comprehensively outlines the pathways of mitochondrial metabolism, emphasizing their critical roles in cellular energy production and metabolic regulation. The associations between aberrant mitochondrial metabolism and the initiation and progression of cancer are highlighted, illustrating how these metabolic disruptions contribute to oncogenesis and tumor sustainability. More importantly, innovative strategies employing nanomedicines to precisely target mitochondrial metabolic pathways in cancer therapy are fully explored. Furthermore, key challenges and future directions in this field are identified and discussed. Collectively, this review provides a comprehensive understanding of the current state and future potential of nanomedicine in targeting mitochondrial metabolism, offering insights for developing more effective cancer therapies.
Collapse
Affiliation(s)
- Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
4
|
Sahoo P, Pathak NK, Scott Bohle D, Dodd EL, Tripathy U. Hematin anhydride (β-hematin): An analogue to malaria pigment hemozoin possesses nonlinearity. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 310:123902. [PMID: 38281463 DOI: 10.1016/j.saa.2024.123902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/08/2023] [Accepted: 01/15/2024] [Indexed: 01/30/2024]
Abstract
Hematin anhydride (β-hematin), the synthetic analogue of the malaria pigment, "hemozoin", is a heme dimer produced by reciprocal covalent bonds among carboxylic acid groups on the protoporphyrin-IX ring and the iron atom present in the two adjacent heme molecules. Hemozoin is a disposal product formed from the digestion of hemoglobin present in the red blood cells infected with hematophagous malaria parasites. Besides, as the parasites invade red blood cells, hemozoin crystals are eventually released into the bloodstream, where they accumulate over time in tissues. Severe malaria infection leads to significant dysfunction in vital organs such as the liver, spleen, and brain in part due to the autoimmune response to the excessive accumulation of hemozoin in these tissues. Also, the amount of these crystals in the vasculature correlates with disease progression. Thus, hemozoin is a unique indicator of infection used as a malaria biomarker and hence, used as a target for the development of antimalarial drugs. Hence, exploring various properties of hemozoin is extremely useful in the direction of diagnosis and cure. The present study focuses on finding one of the unknown properties of β-hematin in physiological conditions by using the Z-scan technique, which is simple, sensitive, and economical. It is observed that hemozoin possesses one of the unique material properties, i.e., nonlinearity with a detection limit of ∼ 15 µM. The self-defocusing action causes β-hematin to exhibit negative refractive nonlinearity. The observed data is analyzed with a thermal lensing model. We strongly believe that our simple and reliable approach to probing the nonlinearity of β-hematin will provide fresh opportunities for malaria diagnostics & cure in the near future.
Collapse
Affiliation(s)
- Priyadarshi Sahoo
- Department of Physics, Indian Institute of Technology (Indian School of Mines), Dhanbad 826004, Jharkhand, India
| | - Nitesh Kumar Pathak
- Department of Physics, Indian Institute of Technology (Indian School of Mines), Dhanbad 826004, Jharkhand, India
| | - D Scott Bohle
- Department of Chemistry, McGill University, Montreal H3A 0B8, Quebec, Canada
| | - Erin L Dodd
- Département de Chimie, Université du Québec à Montréal, 2101, rue Jeanne-Mance Montréal, H2X 2J6 Québec, Canada
| | - Umakanta Tripathy
- Department of Physics, Indian Institute of Technology (Indian School of Mines), Dhanbad 826004, Jharkhand, India.
| |
Collapse
|
5
|
Hu Q, Zhang Y, Mukerabigwi JF, Wang H, Cao Y. Polymer Conjugate as the New Promising Drug Delivery System for Combination Therapy against Cancer. Curr Top Med Chem 2024; 24:1101-1119. [PMID: 39005059 DOI: 10.2174/0115680266280603240321064308] [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: 10/09/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 07/16/2024]
Abstract
This review highlights the advantages of combination therapy using polymer conjugates as drug delivery systems for cancer treatment. In this review, the specific structures and materials of polymer conjugates, as well as the different types of combination chemotherapy strategies, are discussed. Specific targeting strategies, such as monoclonal antibody therapy and small molecule ligands, are also explored. Additionally, self-assembled polymer micelles and overcoming multidrug resistance are described as potential strategies for combination therapy. The assessment of combinational therapeutic efficacy and the challenges associated with polymer conjugates are also addressed. The future outlook aims to overcome these challenges and improve the effectiveness of drug delivery systems for combination therapy. The conclusion emphasizes the potential of polymer conjugates in combination therapy while acknowledging the need for further research and development in this field.
Collapse
Affiliation(s)
- Qiang Hu
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yuannian Zhang
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Jean Felix Mukerabigwi
- Department of Chemistry, University of Rwanda, College of Science and Technology, Po. Box: 3900, Kigali, Rwanda
| | - Haili Wang
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| | - Yu Cao
- Key Laboratory of Pesticide & Chemical Biology (Ministry of Education), National Key Laboratory of Green Pesticide, Engineering Research Center of Photoenergy Utilization for Pollution Control and Carbon Reduction (Ministry of Education), College of Chemistry, Central China Normal University, Wuhan 430079, P.R. China
| |
Collapse
|
6
|
Malekzadeh R, Mortezazadeh T, Abdulsahib WK, Babaye Abdollahi B, Hamblin MR, Mansoori B, Alsaikhan F, Zeng B. Nanoarchitecture-based photothermal ablation of cancer: A systematic review. ENVIRONMENTAL RESEARCH 2023; 236:116526. [PMID: 37487920 DOI: 10.1016/j.envres.2023.116526] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/17/2023] [Accepted: 06/28/2023] [Indexed: 07/26/2023]
Abstract
Photothermal therapy (PTT) is an emerging non-invasive method used in cancer treatment. In PTT, near-infrared laser light is absorbed by a chromophore and converted into heat within the tumor tissue. PTT for cancer usually combines a variety of interactive plasmonic nanomaterials with laser irradiation. PTT enjoys PT agents with high conversion efficiency to convert light into heat to destroy malignant tissue. In this review, published studies concerned with the use of nanoparticles (NPs) in PTT were collected by a systematic and comprehensive search of PubMed, Cochrane, Embase, and Scopus databases. Gold, silver and iron NPs were the most frequent choice in PTT. The use of surface modified NPs allowed selective delivery and led to a precise controlled increase in the local temperature. The presence of NPs during PTT can increase the reactive generation of oxygen species, damage the DNA and mitochondria, leading to cancer cell death mainly via apoptosis. Many studies recently used core-shell metal NPs, and the effects of the polymer coating or ligands targeted to specific cellular receptors in order to increase PTT efficiency were often reported. The effective parameters (NP type, size, concentration, coated polymers or attached ligands, exposure conditions, cell line or type, and cell death mechanisms) were investigated individually. With the advances in chemical synthesis technology, NPs with different shapes, sizes, and coatings can be prepared with desirable properties, to achieve multimodal cancer treatment with precision and specificity.
Collapse
Affiliation(s)
- Reza Malekzadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Medical Radiation Science Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Mortezazadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Waleed K Abdulsahib
- Department of Pharmacology and Toxicology, College of Pharmacy, Al Farahidi University, Baghdad, Iraq
| | - Behnaz Babaye Abdollahi
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Behzad Mansoori
- The Wistar Institute, Cellular and Molecular Oncogenesis Program, Philadelphia, PA, USA.
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Bo Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, 510080, Guangzhou, China.
| |
Collapse
|
7
|
Zhang X, Hu S, Huang L, Chen X, Wang X, Fu YN, Sun H, Li G, Wang X. Advance Progress in Assembly Mechanisms of Carrier-Free Nanodrugs for Cancer Treatment. Molecules 2023; 28:7065. [PMID: 37894544 PMCID: PMC10608994 DOI: 10.3390/molecules28207065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Nanocarriers have been widely studied and applied in the field of cancer treatment. However, conventional nanocarriers still suffer from complicated preparation processes, low drug loading, and potential toxicity of carriers themselves. To tackle the hindrance, carrier-free nanodrugs with biological activity have received increasing attention in cancer therapy. Extensive efforts have been made to exploit new self-assembly methods and mechanisms to expand the scope of carrier-free nanodrugs with enhanced therapeutic performance. In this review, we summarize the advanced progress and applications of carrier-free nanodrugs based on different types of assembly mechanisms and strategies, which involved noncovalent interactions, a combination of covalent bonds and noncovalent interactions, and metal ions-coordinated self-assembly. These carrier-free nanodrugs are introduced in detail according to their assembly and antitumor applications. Finally, the prospects and existing challenges of carrier-free nanodrugs in future development and clinical application are discussed. We hope that this comprehensive review will provide new insights into the rational design of more effective carrier-free nanodrug systems and advancing clinical cancer and other diseases (e.g., bacterial infections) infection treatment.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Shuyang Hu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lifei Huang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiyue Chen
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xin Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ya-nan Fu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hui Sun
- Department of Hepatology, Tongliao Infectious Disease Hospital, Tongliao 028000, China
- Department of Interventional Ultrasound, PLA Medical College & Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
8
|
Hu B, Gao J, Lu Y, Wang Y. Applications of Degradable Hydrogels in Novel Approaches to Disease Treatment and New Modes of Drug Delivery. Pharmaceutics 2023; 15:2370. [PMID: 37896132 PMCID: PMC10610366 DOI: 10.3390/pharmaceutics15102370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 10/29/2023] Open
Abstract
Hydrogels are particularly suitable materials for loading drug delivery agents; their high water content provides a biocompatible environment for most biomolecules, and their cross-linked nature protects the loaded agents from damage. During delivery, the delivered substance usually needs to be released gradually over time, which can be achieved by degradable cross-linked chains. In recent years, biodegradable hydrogels have become a promising technology in new methods of disease treatment and drug delivery methods due to their many advantageous properties. This review briefly discusses the degradation mechanisms of different types of biodegradable hydrogel systems and introduces the specific applications of degradable hydrogels in several new methods of disease treatment and drug delivery methods.
Collapse
Affiliation(s)
- Bo Hu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Jinyuan Gao
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Yu Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China; (B.H.); (J.G.)
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China, Beijing Laboratory of Biomedical Materials, Beijing 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
9
|
Torkamannia A, Omidi Y, Ferdousi R. SYNDEEP: a deep learning approach for the prediction of cancer drugs synergy. Sci Rep 2023; 13:6184. [PMID: 37061563 PMCID: PMC10105711 DOI: 10.1038/s41598-023-33271-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/11/2023] [Indexed: 04/17/2023] Open
Abstract
Drug combinations can be the prime strategy for increasing the initial treatment options in cancer therapy. However, identifying the combinations through experimental approaches is very laborious and costly. Notably, in vitro and/or in vivo examination of all the possible combinations might not be plausible. This study presented a novel computational approach to predicting synergistic drug combinations. Specifically, the deep neural network-based binary classification was utilized to develop the model. Various physicochemical, genomic, protein-protein interaction and protein-metabolite interaction information were used to predict the synergy effects of the combinations of different drugs. The performance of the constructed model was compared with shallow neural network (SNN), k-nearest neighbors (KNN), random forest (RF), support vector machines (SVMs), and gradient boosting classifiers (GBC). Based on our findings, the proposed deep neural network model was found to be capable of predicting synergistic drug combinations with high accuracy. The prediction accuracy and AUC metrics for this model were 92.21% and 97.32% in tenfold cross-validation. According to the results, the integration of different types of physicochemical and genomics features leads to more accurate prediction of synergy in cancer drugs.
Collapse
Affiliation(s)
- Anna Torkamannia
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, 51656/65811, Iran
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA
| | - Reza Ferdousi
- Department of Health Information Technology, School of Management and Medical Informatics, Tabriz University of Medical Sciences, Tabriz, 51656/65811, Iran.
| |
Collapse
|
10
|
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
|
11
|
Sarikhani A, Alamzadeh Z, Beik J, Irajirad R, Mirrahimi M, Pirhajati Mahabadi V, Kamrava SK, Ghaznavi H, Khoei S. Ultrasmall Fe3O4 and Gd2O3 hybrid nanoparticles for T1-weighted MR imaging of cancer. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00148-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AbstractGadolinium-based contrast agents (GdCAs) have been the most frequently used T1-weighted magnetic resonance imaging (MRI) contrast agents for decades. However, they have serious disadvantages such as low longitudinal relaxivity value (r1) and high dose associated-nephrotoxicity that restrict their wide applications. These emphasize the need for an ideal stable and biocompatible T1-weighted CA with high contrast enhancement performance. Here, we propose a wet-chemical synthesis technique to form a nanocomposite consisting of ultrasmall iron oxide nanoparticles (US-IO) and Gd2O3 hybrid nanoparticles stabilized with dextran (FG-HNPs) for T1-weighted MR imaging. Relaxometry study showed that FG-HNPs have a high r1 value (42.28 mM−1S−1) and low relaxivity ratio (r2/r1: 1.416) at 3.0T. In vivo MRI contrast enhancement factor (ΔSNR) for FG-HNPs (257.025 ± 17.4%) was found to be 1.99-fold higher than US-IO (129.102 ± 15%) and 3.35-fold higher than Dotarem (76.71 ± 14.2%) as routinely used T1-weighted CA. The cytotoxicity assay and histological examination confirmed the biocompatibility of FG-HNPs. The biodistribution study, transmission electron microscopy (TEM) and Prussian blue (PB) staining of tumor tissue proved the effective tumor localization of FG-HNPs. Therefore, FG-HNPs can be suggested as a promising CA for T1-weighted MRI of tumors by virtue of their remarkable relaxivities and high biocompatibility.
Collapse
|
12
|
Ali AA, Abuwatfa WH, Al-Sayah MH, Husseini GA. Gold-Nanoparticle Hybrid Nanostructures for Multimodal Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12203706. [PMID: 36296896 PMCID: PMC9608376 DOI: 10.3390/nano12203706] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/12/2022] [Accepted: 10/18/2022] [Indexed: 06/01/2023]
Abstract
With the urgent need for bio-nanomaterials to improve the currently available cancer treatments, gold nanoparticle (GNP) hybrid nanostructures are rapidly rising as promising multimodal candidates for cancer therapy. Gold nanoparticles (GNPs) have been hybridized with several nanocarriers, including liposomes and polymers, to achieve chemotherapy, photothermal therapy, radiotherapy, and imaging using a single composite. The GNP nanohybrids used for targeted chemotherapy can be designed to respond to external stimuli such as heat or internal stimuli such as intratumoral pH. Despite their promise for multimodal cancer therapy, there are currently no reviews summarizing the current status of GNP nanohybrid use for cancer theragnostics. Therefore, this review fulfills this gap in the literature by providing a critical analysis of the data available on the use of GNP nanohybrids for cancer treatment with a specific focus on synergistic approaches (i.e., triggered drug release, photothermal therapy, and radiotherapy). It also highlights some of the challenges that hinder the clinical translation of GNP hybrid nanostructures from bench to bedside. Future studies that could expedite the clinical progress of GNPs, as well as the future possibility of improving GNP nanohybrids for cancer theragnostics, are also summarized.
Collapse
Affiliation(s)
- Amaal Abdulraqeb Ali
- Biomedical Engineering Graduate Program, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Mohammad H. Al-Sayah
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
13
|
Xie X, Jiang K, Li B, Hou S, Tang H, Shao B, Ping Y, Zhang Q. A small-molecule self-assembled nanodrug for combination therapy of photothermal-differentiation-chemotherapy of breast cancer stem cells. Biomaterials 2022; 286:121598. [DOI: 10.1016/j.biomaterials.2022.121598] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 02/07/2023]
|
14
|
Ghalandari B, Asadollahi K, Ghorbani F, Ghalehbaghi S, Rafiee S, Komeili A, Kamrava SK. Determinants of gold nanoparticle interactions with Proteins: Off-Target effect study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 269:120736. [PMID: 34923375 DOI: 10.1016/j.saa.2021.120736] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/17/2021] [Accepted: 12/07/2021] [Indexed: 06/14/2023]
Abstract
Photothermal therapy is one of the promising approaches toward cancer treatment. To date, several compounds have been developed for this application, among which nanoparticles are attracting ever-increasing attention. One of the obstacles in developing efficient photothermal nanoparticle agents is their off-target effect which is mainly mediated via non-specific interactions with proteins. Such interaction not only reduces the bioavailability of the agent but also will cause protein aggregation that can be lethal. So, gaining knowledge on the mechanisms mediating such interactions will facilitate development of more effective agents. Our last studies showed the mechanism of action of two modified gold nanoparticles, folic acid functionalized gold nanoparticles (FA-AuNPs) and gold shelled Fe3O4 nanoparticles (AuFeNPs), as photothermal agents. In the current work, we focus on the interaction of these two NPs with human serum albumin (HSA) and human hemoglobin (Hb) as model proteins. The complex formation between NPs and proteins was investigated by fluorescence spectroscopy, dynamic light scattering and circular dichroism. Our data distinguishes the very distinct mode of interaction of charged and neutral NPs with proteins. While the interaction of neutral AuFeNP to proteins is protein dependent, charged nanoparticles FA-AuNP interact indistinguishably with all proteins via electrostatic interactions. Moreover, complexes obtained from FA-AuNPs with proteins are more stable than that of AuFeNP. However, the secondary structure content of proteins in the presence of NPs indicates the insignificant effect of NPs on the secondary structure of these proteins. Our data propose that the charge functionalization of the NPs is an effective way for modulating the interaction of nanoparticles with proteins.
Collapse
Affiliation(s)
- Behafarid Ghalandari
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Kazem Asadollahi
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Farnaz Ghorbani
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai 201399, China
| | - Suzan Ghalehbaghi
- Medical Engineering Department, South Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Saharnaz Rafiee
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ali Komeili
- Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Kamran Kamrava
- Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
15
|
Nonlinear optical response of cancer cells following conventional and nano-technology based treatment strategies: Results of chemo-, thermo- and radiation therapies. Photodiagnosis Photodyn Ther 2021; 37:102686. [PMID: 34915185 DOI: 10.1016/j.pdpdt.2021.102686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/30/2021] [Accepted: 12/10/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Although traditional treatments are able to increase cancer survival rate, undesirable impact on off-target tissues are considered a limitation of these approaches. Nanotechnology-based treatments have been proposed as a possible option to enhance targeting., Further,current methods for evaluating cellular damage, are time consuming, highly dependent on the operator skills, and expensive. The aim of this study was to evaluate the capability of nonlinear optical response of cells to determine cellular damages during conventional and nano-technology based treatments. METHODS Three different cancer cell lines, CT26, KB, and MCF-7 were used in this study. The alginate hydrogel co-loaded with cisplatin and Au nanoparticle (ACA) nanocomplex and gold-coated iron oxide nanoparticle (Au@IONP) were considered for chemo- and chemo-photothermal therapies, and thermo-radiation therapy, respectively. The sign and value of nonlinear optical absorption coefficient and imaginary part of the third-order nonlinear susceptibility of cells were computed. MTT assay was utilized as a reference method. RESULTS The value of nonlinear optical indices increased with increasing cellular damage and cell death. The linear regression analysis indicated high correlation between nonlinear optical indices and MTT results, in all treatments. CONCLUSION The nonlinear optical indices are robust from confounding factors, namely treatment approach (traditional and nano-technology based), treatment modality (chemotherapy, thermotherapy, photothermal therapy, and radiation therapy), and cell types. Nonlinear optical properties of cells can be used as a rapid estimation method for cell damage, at the nanoscale level.
Collapse
|
16
|
Beik J, Alamzadeh Z, Mirrahimi M, Sarikhani A, Ardakani TS, Asadi M, Irajirad R, Mirrahimi M, Mahabadi VP, Eslahi N, Bulte JWM, Ghaznavi H, Shakeri-Zadeh A. Multifunctional Theranostic Graphene Oxide Nanoflakes as MR Imaging Agents with Enhanced Photothermal and Radiosensitizing Properties. ACS APPLIED BIO MATERIALS 2021; 4:4280-4291. [PMID: 35006840 DOI: 10.1021/acsabm.1c00104] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The integration of multiple therapeutic and diagnostic functions into a single nanoplatform for image-guided cancer therapy has been an emerging trend in nanomedicine. We show here that multifunctional theranostic nanostructures consisting of superparamagnetic iron oxide (SPIO) and gold nanoparticles (AuNPs) scaffolded within graphene oxide nanoflakes (GO-SPIO-Au NFs) can be used for dual photo/radiotherapy by virtue of the near-infrared (NIR) absorbance of GO for photothermal therapy (PTT) and the Z element radiosensitization of AuNPs for enhanced radiation therapy (RT). At the same time, this nanoplatform can also be detected by magnetic resonance (MR) imaging because of the presence of SPIO NPs. Using a mouse carcinoma model, GO-SPIO-Au NF-mediated combined PTT/RT exhibited a 1.85-fold and 1.44-fold higher therapeutic efficacy compared to either NF-mediated PTT or RT alone, respectively, resulting in a complete eradication of tumors. As a sensitive multifunctional theranostic platform, GO-SPIO-Au NFs appear to be a promising nanomaterial for enhanced cancer imaging and therapy.
Collapse
Affiliation(s)
- Jaber Beik
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Alamzadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehri Mirrahimi
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Sarikhani
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mohamadreza Asadi
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Irajirad
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehraban Mirrahimi
- Biology Department, School of Science, Tehran University of Medical Sciences, 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
| | - Neda Eslahi
- Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Jeff W M Bulte
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ali Shakeri-Zadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Liu F, Wang X, Liu Q, Zhang H, Xie L, Wang Q, Li L, Li R. Biocompatible Nanoparticles as a Platform for Enhancing Antitumor Efficacy of Cisplatin-Tetradrine Combination. NANOSCALE RESEARCH LETTERS 2021; 16:61. [PMID: 33855646 PMCID: PMC8046896 DOI: 10.1186/s11671-021-03511-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 03/21/2021] [Indexed: 06/12/2023]
Abstract
Combination therapy has been a standard strategy in the clinical tumor treatment. We have demonstrated that combination of Tetradrine (Tet) and Cisplatin (CDDP) presented a marked synergistic anticancer activity, but inevitable side effects limit their therapeutic concentration. Considering the different physicochemical and pharmacokinetic properties of the two drugs, we loaded them into a nanovehicle together by the improved double emulsion method. The nanoparticles (NPs) were prepared from the mixture of poly(ethyleneglycol)-polycaprolactone (PEG-PCL) and polycarprolactone (HO-PCL), so CDDP and Tet can be located into the NPs simultaneously, resulting in low interfering effect and high stability. Images from fluorescence microscope revealed the cellular uptake of both hydrophilic and hydrophobic agents delivered by the NPs. In vitro studies on different tumor cell lines and tumor tissue revealed increased tumor inhibition and apoptosis rates. As to the in vivo studies, superior antitumor efficacy and reduced side effects were observed in the NPs group. Furthermore, 18FDG-PET/CT imaging demonstrated that NPs reduced metabolic activities of tumors more prominently. Our results suggest that PEG-PCL block copolymeric NPs could be a promising carrier for combined chemotherapy with solid efficacy and minor side effects.
Collapse
Affiliation(s)
- Fangcen Liu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xinyue Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qin Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Huan Zhang
- Center for Personalized Medicine, Linköping University, 58183, Linköping, Sweden
| | - Li Xie
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qin Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rutian Li
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
18
|
Saatchian E, Ehsani S, Sarikhani A, Ghaznavi H, Montazerabadi A, Shakeri-Zadeh A. Monitoring of the choline/lipid ratio by 1H-MRS can be helpful for prediction and early detection of tumor response to nano-photo-thermal therapy. Lasers Med Sci 2021; 37:335-343. [PMID: 33523392 DOI: 10.1007/s10103-021-03256-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022]
Abstract
Nanotechnology-based photothermal therapy (NPTT) is a new emerging modality of cancer therapy. To have the right prediction and early detection of response to NPTT, it is necessary to get rapid feedback from a tumor treated by NPTT procedure and stay informed of what happens in the tumor site. We performed this study to find if proton magnetic resonance spectroscopy (1H-MRS) can be well responsive to such an imperative requirement. We considered various treatment groups including gold nanoparticles (AuNPs), laser, and the combination of AuNPs and laser (NPTT group). Therapeutic effects on CT26 colon tumor-bearing BALB/c mice were studied by looking at alterations that happened in 1H-MRS signals and tumor size after conducting treatment procedures. In MRS studies, the alterations of choline and lipid concentrations and their ratio were investigated. Having normalized the metabolite peak to water peak, we found a significant decrease in choline concentration post-NPTT (from (1.25 ± 0.05) × 10-3 to (0.43 ± 0.04) × 10-3), while the level of lipid concentration in the tumor was slightly increased (from (2.91 ± 0.23) × 10-3 to (3.52 ± 0.31) × 10-3). As a result, the choline/lipid ratio was significantly decreased post-NPTT (from 0.41 ± 0.11 to 0.11 ± 0.02). Such alterations appeared just 1 day after NPTT. Tumor shrinkage in all groups was studied and significant changes were significantly detectable on day 7 post-NPTT procedure. In conclusion, the study of choline/lipid ratio using 1H-MRS may help us estimate what happens in a tumor treated by the NPTT method. Such an in vivo assessment is interestingly feasible as soon as just 1 day post-NPTT. This would undoubtedly help the oncologists make a more precise decision about treatment planning strategies. Monitoring of the choline/lipid ratio by 1H-MRS can be helpful for prediction and early detection of response to nano-photo-thermal therapy.
Collapse
Affiliation(s)
- Erfan Saatchian
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sina Ehsani
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Sarikhani
- Finetech in Medicine Research Center, Iran University of Medical Science, Tehran, Iran
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences (ZaUMS), Zahedan, Iran.
| | - Alireza Montazerabadi
- Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Shakeri-Zadeh
- Finetech in Medicine Research Center, Iran University of Medical Science, Tehran, Iran
| |
Collapse
|
19
|
Afzali E, Eslaminejad T, Yazdi Rouholamini SE, Shahrokhi-Farjah M, Ansari M. Cytotoxicity Effects of Curcumin Loaded on Chitosan Alginate Nanospheres on the KMBC-10 Spheroids Cell Line. Int J Nanomedicine 2021; 16:579-589. [PMID: 33531802 PMCID: PMC7846832 DOI: 10.2147/ijn.s251056] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose Breast cancer is one of the most lethal types of cancer in women. Curcumin showed therapeutic potential against breast cancer, but applying that by itself does not lead to the associated health benefits due to its poor bioavailability, which appears to be primarily due to poor absorption, rapid metabolism, and rapid elimination. Moreover, poor water solubility of curcumin causes accumulation of a high concentration of curcumin and so decrease its permeability to the cell. Many strategies are employed to reduce curcumin metabolism such as adjuvants and designing novel delivery systems. Therefore, in this study sodium alginate and chitosan were used to synthesize the hydrogels that are known as biocompatible, hydrophilic and low toxic drug delivery systems. Also, folic acid was used to link to chitosan in order to actively targetfolate receptors on the cells. Methods Chitosan-β-cyclodextrin-TPP-Folic acid/alginate nanoparticles were synthesized and then curcumin was loaded on them. Interaction between the constituents of the particles was characterized by FTIR spectroscopy. Morphological structures of samples were studied by FE-SEM. Release profile of curcumin was determined by dialysis membrane. The cytotoxic test was done on the Kerman male breast cancer (KMBC-10) cell line by using MTT assay. The viability of cells was detected by fluorescent staining. Gene expression was investigated by real-time PCR. Results The encapsulation of curcumin into nano-particles showed an almost spherical shape and an average particle size of 155 nm. In vitro cytotoxicity investigation was indicated as dose-respond reaction against cancer breast cells after 24 h incubation. On the other hand, in vitro cell uptake study revealed active targeting of CUR-NPs into spheroids. Besides, CXCR4 expression was detected about 30-fold less than curcumin alone. The CUR-NPs inhibited proliferation and increased apoptosis in spheroid human breast cancer cells. Conclusion Our results showed the potential of NPs as an effective candidate for curcumin delivery to the target tumor spheroids that confirmed the creatable role of folate receptors.
Collapse
Affiliation(s)
- Elham Afzali
- Pharmaceutics Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Touba Eslaminejad
- Pharmaceutics Research Centre, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyede Elmira Yazdi Rouholamini
- Physiology Research Centre, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mariam Shahrokhi-Farjah
- Physiology Research Centre, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mehdi Ansari
- Department of Drug and Food Control, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
20
|
Wang L, Tang S, Yu Y, Lv Y, Wang A, Yan X, Li N, Sha C, Sun K, Li Y. Intranasal Delivery of Temozolomide-Conjugated Gold Nanoparticles Functionalized with Anti-EphA3 for Glioblastoma Targeting. Mol Pharm 2021; 18:915-927. [PMID: 33417456 DOI: 10.1021/acs.molpharmaceut.0c00911] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly lethal and aggressive tumor of the brain that carries a poor prognosis. Temozolomide (TMZ) has been widely used as a first-line treatment for GBM. However, poor brain targeting, side effects, and drug resistance limit its application for the treatment of GBM. We designed a Temozolomide-conjugated gold nanoparticle functionalized with an antibody against the ephrin type-A receptor 3 (anti-EphA3-TMZ@GNPs) for targeted GBM therapy via intranasal administration. The system can bypass the blood-brain barrier and target active glioma cells to improve the glioma targeting of TMZ and enhance the treatment efficacy, while reducing the peripheral toxicity and drug resistance. The prepared anti-EphA3-TMZ@GNPs were 46.12 ± 2.0 nm and suitable for intranasal administration, which demonstrated high safety to the nasal mucosa in a toxicity assay. In vitro studies showed that anti-EphA3-TMZ@GNPs exhibited significantly enhanced cellular uptake and toxicity, and a higher cell apoptosis ratio has been seen compared with that of TMZ (54.9 and 14.1%, respectively) toward glioma cells (C6). The results from experiments on TMZ-resistant glioma cells (T98G) demonstrated that the IC50 of anti-EphA3-TMZ@GNPs (64.06 ± 0.16 μM) was 18.5-fold lower than that of TMZ. In addition, Western blot analysis also revealed that anti-EphA3-TMZ@GNPs effectively down-modulated expression of O6-methylguanine-DNA methyltransferase and increased chemosensitivity of T98G to TMZ. The antiglioma efficacy in vivo was investigated in orthotopic glioma-bearing rats, and the results demonstrated that the anti-EphA3-TMZ@GNPs prolonged the median survival time to 42 days and increased tumor-cell apoptosis dramatically compared with TMZ. In conclusion, anti-EphA3-TMZ@GNPs could serve as an intranasal drug delivery system for efficacious treatment of GBM.
Collapse
Affiliation(s)
- Liangxiao Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Shengnan Tang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Yawen Yu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Yanan Lv
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Aiping Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Xiuju Yan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Nuannuan Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Chunjie Sha
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Luye Pharmaceutical Co., Ltd., Yantai 264003, P.R. China
| | - Kaoxiang Sun
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| | - Youxin Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, P.R. China
| |
Collapse
|
21
|
Huang L, Zhao S, Fang F, Xu T, Lan M, Zhang J. Advances and perspectives in carrier-free nanodrugs for cancer chemo-monotherapy and combination therapy. Biomaterials 2020; 268:120557. [PMID: 33260095 DOI: 10.1016/j.biomaterials.2020.120557] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/09/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Nanocarrier-based drug delivery systems hold impressive promise for biomedical application because of their excellent water dispersibility, prolonged blood circulation time, increased drug accumulation in tumors, and potential in combination therapeutics. However, most nanocarriers suffer from low drug-loading efficiency, poor therapeutic effectiveness, potential systematic toxicity, and unstable metabolism. As an alternative, carrier-free nanodrugs, completely formulated with one or more drugs, have attracted increasing attention in cancer therapy due to their advantage of improved pharmacodynamics/pharmacokinetics, reduced toxicity, and high drug-loading. In recent years, carrier-free nanodrugs have contributed to progress in a variety of therapeutic modalities. In this review, different common strategies for carrier-free nanodrugs preparation are first summarized, mainly including nanoprecipitation, template-assisted nanoprecipitation, thin-film hydration, spray-drying technique, supercritical fluid (SCF) technique, and wet media milling. Then we describe the recently reported carrier-free nanodrugs for cancer chemo-monotherapy or combination therapy. The advantages of anti-cancer drugs combined with other chemotherapeutic, photosensitizers, photothermal, immunotherapeutic or gene drugs have been demonstrated. Finally, a future perspective is introduced to highlight the existing challenges and possible solutions toward clinical application of currently developed carrier-free nanodrugs, which may be instructive to the design of effective carrier-free regimens in the future.
Collapse
Affiliation(s)
- Li Huang
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China
| | - Shaojing Zhao
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China
| | - Fang Fang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Ting Xu
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China
| | - Minhuan Lan
- Hunan Provincial Key Laboratory of Micro & Nano Materials Interface Science, College of Chemistry and Chemical Engineering, Central South University, Changsha, 410083, PR China.
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Sciences, Beijing Institute of Technology, Beijing, 100081, PR China.
| |
Collapse
|
22
|
Xu P, Liang F. Nanomaterial-Based Tumor Photothermal Immunotherapy. Int J Nanomedicine 2020; 15:9159-9180. [PMID: 33244232 PMCID: PMC7684030 DOI: 10.2147/ijn.s249252] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, photothermal therapy (PTT) particularly nanomaterial-based PTT is a promising therapeutic modality and technique for cancer tumor ablation. In addition to killing tumor cells directly through heat, PTT also can induce immunogenic cell death (ICD) to activate the whole-body anti-tumor immune response, including the redistribution and activation of immune effector cells, the expression and secretion of cytokines and the transformation of memory T lymphocytes. When used in combination with immunotherapy, the efficacy of nanomaterial-based PTT can be improved. This article summarized the mechanism of nanomaterial-based PTT against cancer and how nanomaterial-based PTT impacts the tumor microenvironment and induces an immune response. Moreover, we reviewed recent advances of nanomaterial-based photothermal immunotherapy and discussed challenges and future outlook.
Collapse
Affiliation(s)
- Peng Xu
- The State Key Laboratory of Refractories and Metallurgy, Coal Conversion and New Carbon Materials Hubei Key Laboratory, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan430081, People’s Republic of China
| | - Feng Liang
- The State Key Laboratory of Refractories and Metallurgy, Coal Conversion and New Carbon Materials Hubei Key Laboratory, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan430081, People’s Republic of China
| |
Collapse
|
23
|
Chen H, Wang X, Sutrisno L, Zeng T, Kawazoe N, Yang Y, Chen G. Folic Acid-Functionalized Composite Scaffolds of Gelatin and Gold Nanoparticles for Photothermal Ablation of Breast Cancer Cells. Front Bioeng Biotechnol 2020; 8:589905. [PMID: 33251201 PMCID: PMC7671968 DOI: 10.3389/fbioe.2020.589905] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/24/2020] [Indexed: 12/24/2022] Open
Abstract
Photothermal therapy (PTT) has been developed as a useful therapeutic method for cancer treatment. Localization of PTT agents in cancer sites and targeting capacity are required to further increase therapeutic efficacy. In this study, gold nanoparticles (AuNPs) and gelatin were functionalized with folic acid (FA) and hybridized to prepare FA-functionalized gelatin-AuNPs composite scaffolds. AuNPs with rod and star shapes of three sizes (40, 70, and 110 nm) were used for the hybridization to investigate the influence of AuNPs shape and size. The composite scaffolds showed porous structures with good interconnectivity. Modification with FA increased capture capacity of the composite scaffolds. Hybridization with AuNPs rendered the composite scaffold a good photothermal conversion property under near-infrared (NIR) laser irradiation. Temperature change during laser irradiation increased with the laser power intensity and irradiation time. The shape and size of AuNPs also affected their photothermal conversion property. The composite scaffold of gold nanorods 70 (FA-G/R70) had the highest photothermal conversion capacity. Breast cancer cells cultured in the FA-G/R70 composite scaffold were killed under NIR laser irradiation. Mouse subcutaneous implantation further demonstrated the excellent photothermal ablation capability of FA-G/R70 composite scaffold to breast cancer cells. The FA-functionalized composite scaffolds were demonstrated a high potential for local PPT of breast cancer.
Collapse
Affiliation(s)
- Huajian Chen
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan.,Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Xiuhui Wang
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan.,Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Linawati Sutrisno
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan.,Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tianjiao Zeng
- Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan
| | - Yingnan Yang
- Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan.,Department of Materials Science and Engineering, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
24
|
Liu TI, Lu TY, Yang YC, Chang SH, Chen HH, Lu IL, Sabu A, Chiu HC. New combination treatment from ROS-Induced sensitized radiotherapy with nanophototherapeutics to fully eradicate orthotopic breast cancer and inhibit metastasis. Biomaterials 2020; 257:120229. [DOI: 10.1016/j.biomaterials.2020.120229] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 02/06/2023]
|
25
|
Liu J, Zhang Y, Li Q, Feng Z, Huang P, Wang W, Liu J. Development of injectable thermosensitive polypeptide hydrogel as facile radioisotope and radiosensitizer hotspot for synergistic brachytherapy. Acta Biomater 2020; 114:133-145. [PMID: 32688087 DOI: 10.1016/j.actbio.2020.07.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 01/02/2023]
Abstract
Brachytherapy is considered to be an unparalleled form of conformal radiation therapy, which involves the delivery of radiation directly to tumor lesions or the postoperative cavity. With the development of specific applicators, the exploitation of in situ drug-delivery platform introduces opportunities for the synchronous administration of radiosensitizers. In this study, an iodine-131 (I131)-labeled injectable thermosensitive methoxy poly(ethylene glycol)-b-poly(tyrosine) hydrogel (denoted as PETyr-I131) was developed via a facile method. The radioactive source of I131 was immobilized at the subcutaneous injection site and monitored via single-photon emission computed tomography in real time, and hematological and histopathological analyses revealed no obvious side effects. Additionally, the SmacN7 peptide conjugated with cell membrane-permeable oligosarginine (denoted as SmacN7-R9) was used to enhance the radiosensitivity of cancer cells, as confirmed by the results of reactive oxygen species detection, DNA damage assay, cell apoptosis assay, and clonogenic evaluation. Importantly, a synergistic brachytherapy treatment effect on tumor-bearing nude mice was achieved. The proposed thermosensitive supramolecular hydrogel platform, which conformally immobilizes radionuclides and delivers radiosensitizers by virtue of its proximity to the site of the primary tumor or the postoperative cavity, has great potential for achieving synergistic treatment outcomes with reduced radiation-related side effects. STATEMENT OF SIGNIFICANCE: In this work, a kind of radioiodinated thermosensitive supramolecular hydrogel was developed, which was facilely used as the radioactive source for brachytherapy. Meanwhile, SmacN7-R9 peptide was combined as a model radiosensitizer to facilitate the activation of tumor cell apoptosis pathways and promotion of radiation-induced cytotoxicity. Synergistic brachytherapy outcomes were achieved from the in vitro and in vivo evaluations. Therefore, from the practical standpoint, this thermosensitive supramolecular hydrogel platform holds great potential for the 3D-conformally immobilizing radionuclide and delivering radiosensitizer by virtue of its proximity to the site of primary tumor lesions or postoperative cavity, resulting in synergetic treatment outcomes with reduced radiation associated side effects.
Collapse
|
26
|
Xie Z, Lu R, Zhu Y, Peng M, Fan T, Ren P, Wang B, Kang L, Liu X, Li S, Cui H. Liquid-phase exfoliation of black sesame to create a nanoplatform for in vitro photoluminescence and photothermal therapy. Nanomedicine (Lond) 2020; 15:2041-2052. [PMID: 32867583 DOI: 10.2217/nnm-2020-0151] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The present study aims to apply the facile liquid-phase exfoliation (LPE) strategy to fabricate 2D organic materials and thus to broaden the family of biocompatible and multifunctional 2D materials. Materials & methods: 2D material-organic melanin and cellulose nanosheets were synthesized from black sesame hull using LPE. Photoluminescence and photothermal properties of the nanosheets were assessed, as well as stability and cell killing ability. Results: The prepared 2D nanoplatform exhibited broad and multiple photoluminescent emission bands. It also demonstrated efficient photothermal cancer therapy with excellent biocompatibility. Conclusion: The present study could open an avenue in exfoliating organic materials using the LPE strategy. This could make the fabrication of multifunctional 2D organic materials more efficient and broaden the family of biocompatible 2D nanomaterials.
Collapse
Affiliation(s)
- Zhongjian Xie
- Shenzhen International Institute for Biomedical Research, Shenzhen, Guangdong, 518116, PR China
| | - Ruitao Lu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, Guangdong, PR China
| | - Yao Zhu
- Department of Ultrasonography, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science & Technology, Shenzhen, Guangdong, 518020, PR China
| | - Minhua Peng
- Shenzhen International Institute for Biomedical Research, Shenzhen, Guangdong, 518116, PR China.,Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, PR China
| | - Taojian Fan
- Key Laboratory of Optoelectronic Devices & Systems of Ministry of Education & Guangdong Province, Institute of Microscale Optoelectronics, & Otolaryngology Department & Biobank of the First Affiliated Hospital, Shenzhen Second People's Hospital, Health Science Center, Shenzhen University, Shenzhen, 518060, PR China
| | - Peigen Ren
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, PR China
| | - Bing Wang
- College of Physics & Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, PR China
| | - Lin Kang
- Clinical Medical Research Center, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518020, Guangdong, PR China.,Department of Endocrinology, Shenzhen People's Hospital, Shenzhen, 518020, Guangdong, PR China
| | - Xiaoyun Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, 215 West Heping Road, Shijiazhuang, Hebei, 050000, PR China
| | - Sha Li
- Department of Anatomy, Hebei Medical University, Shijiazhuang, PR China
| | - Huixian Cui
- Department of Anatomy, Hebei Medical University, Shijiazhuang, PR China
| |
Collapse
|
27
|
Amani S, Mehdizadeh A, Movahedi MM, Keshavarz M, Koosha F. Investigation of the Dose-Enhancement Effects of Spherical and Rod-Shaped Gold Nanoparticles on the HeLa Cell Line. Galen Med J 2020; 9:e1581. [PMID: 34466556 PMCID: PMC8343815 DOI: 10.31661/gmj.v9i0.1581] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 05/31/2020] [Accepted: 06/15/2020] [Indexed: 01/05/2023] Open
Abstract
Background: Cervical cancer cells are known as radioresistant cells. Current treatment methods have not improved the patients’ survival efficiently; thus, new therapeutic strategies are needed to enhance the efficacy of radiotherapy. Gold nanomaterials with different shapes and sizes have been explored as radiosensitizers. The present study compared the radiosensitizing effects of gold nanorods (AuNRs) with spherical gold nanoparticles (AuNPs) on the HeLa cell line irradiated with megavoltage X-rays. Materials and Methods: The cytotoxicity of AuNRs and AuNPs on HeLa cells in the presence and absence of 6-MV X-ray was investigated using the MTT assay. For this aim, HeLa cells were incubated with and AuNPs and AuNRs at various concentrations (5, 10, and 15 µg/mL) for 6 hours. Afterward, HeLa cells were irradiated with 6-MV X-ray at a single dose of 2 Gy. Results: The results showed that the addition of AuNRs and AuNPs could enhance the radiosensitivity of HeLa cells. Both AuNRs and AuNPs showed low toxicity on HeLa cells, while AuNRs were more toxic than AuNPs at the examined concentrations. Moreover, it was found that AuNRs could enhance the radiosensitivity of HeLa cells more than spherical-shaped AuNPs. Conclusion: This study revealed that the shape of nanoparticles is an effective factor when they are used as radiosensitizing agents during radiotherapy.
Collapse
Affiliation(s)
- Samad Amani
- Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Mehdizadeh
- Department of Medical Physics and Medical Engineering, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Mehdi Movahedi
- Department of Medical Physics and Medical Engineering, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Marzieh Keshavarz
- Department of Medical Physics and Medical Engineering, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Ionizing and Non-ionizing Radiation Protection Research Center (INIRPRC), Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Koosha
- Department of Radiology Technology, Faculty of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Correspondence to: Fereshteh Koosha, Ph.D, Assistant Professor, Department of Radiology Technology, Faculty of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Darband St, Ghods Sq., Tehran, Iran Telephone Number: +98-2122717503 Email Address:
| |
Collapse
|
28
|
Mahajan S, Raval N, Kalyane D, Anup N, Maheshwari R, Tambe V, Kalia K, Tekade RK. NanoGold-core dendrimeric seeds for combined chemo-, photothermal-, and photodynamic therapy of cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101814] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
29
|
Xie G, Zhang L, Pan J, Zhang X, Sun SK. Green and Kilogram-Scale Synthesis of Fe Hydrogel for Photothermal Therapy of Tumors in Vivo. ACS Biomater Sci Eng 2020; 6:4276-4284. [PMID: 33463327 DOI: 10.1021/acsbiomaterials.9b01933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Photothermal agents with good biocompatibility, high tumor accumulation efficiency, large-scale production ability, and low cost are crucial for potential photothermal treatment in clinic. Herein, we proposed a green and highly efficient strategy to fabricate a kilogram-scale alginate-Ca2+-Fe powder hydrogel (ALG-Ca2+-Fe) by turning commercial Fe powder into hydrogel for enhanced photothermal therapy. The ALG-Ca2+-Fe was formed by simply dispersing commercial Fe powder into the preformed alginate-Ca2+ hydrogel in a green and energy-/time-saving way. The hydrogel exhibited the advantages of ultrahigh loading capacity of Fe powder (>100 mg mL-1), excellent large-scale production capacity (>1 kg in lab synthesis), low cost (<1.7 $/kg), and good injectability. More importantly, large size and hydrophobicity endowed Fe powder with excellent tumor retention effect and minimal diffusion to surrounding tissues, greatly benefiting improving treatment efficiency and reducing side effects. In vivo and in vitro studies both proved that the large-scale produced ALG-Ca2+-Fe can be used for highly efficient and biosafe tumor treatment in vivo by simple noninvasive injection. The developed ALG-Ca2+-Fe with multiple superiors opens up a novel green way to develop efficient and safe photothermal therapeutic agents with great clinic transformation potential.
Collapse
Affiliation(s)
- Guangchao Xie
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Liang Zhang
- Department of Ultrasound, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xuejun Zhang
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| | - Shao-Kai Sun
- School of Medical Imaging, Tianjin Medical University, Tianjin 300203, China
| |
Collapse
|
30
|
Tabei M, Zeinizade E, Beik J, Kamrava SK, Nasiri Z, Ghaznavi H, Shakeri-Zadeh A. Insights into Nano-Photo-Thermal Therapy of Cancer: The Kinetics of Cell Death and Effect on Cell Cycle. Anticancer Agents Med Chem 2020; 20:612-621. [DOI: 10.2174/1871520620666200129111332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 10/31/2019] [Accepted: 11/12/2019] [Indexed: 11/22/2022]
Abstract
Background:
Despite considerable advances in nano-photo-thermal therapy (NPTT), there have been
a few studies reporting in-depth kinetics of cell death triggered by such a new modality of cancer treatment.
Objective:
In this study, we aimed to (1) investigate the cell death pathways regulating the apoptotic responses
to NPTT; and (2) ascertain the effect of NPTT on cell cycle progression.
Methods:
Folate conjugated gold nanoparticle (F-AuNP) was firstly synthesized, characterized and then assessed
to determine its potentials in targeted NPTT. The experiments were conducted on KB nasopharyngeal
cancer cells overexpressing folate receptors (FRs), as the model, and L929 normal fibroblast cells with a low
level of FRs, as the control. Cytotoxicity was evaluated by MTT assay and the cell death mode (i.e., necrosis or
apoptosis) was determined through AnnexinV/FITC-propidium iodide staining. Next, the gene expression profiles
of some key apoptotic factors involved in the mitochondrial signaling pathway were investigated using
RT-qPCR. Finally, cell cycle phase distribution was investigated at different time points post NPTT using flow
cytometric analysis.
Results:
The obtained results showed that KB cell death following targeted NPTT was greater than that observed
for L929 cells. The majority of KB cell death following NPTT was related to apoptosis. RT-qPCR analysis
indicated that the elevated expression of Bax along with the depressed expression of Bcl-xL, Survivin and
XIAP may involve in the regulation of apoptosis in response to NPTT. Flow cytometric analysis manifested that
16-24 hours after NPTT, the major proportion of KB cells was in the most radiosensitive phases of the cell cycle
(G2/M).
Conclusion:
This study extended the understanding of the signaling pathway involved in the apoptotic response
to NPTT. Moreover, the potential effect of NPTT on sensitizing cancer cells to subsequent radiation therapy was
highlighted.
Collapse
Affiliation(s)
- Mousa Tabei
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Elham Zeinizade
- 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
| | - S. Kamran Kamrava
- ENT and Head & Neck Research Center and Department, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Zahra Nasiri
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Habib Ghaznavi
- Department of Pharmaceutical Research Centre, 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
| |
Collapse
|
31
|
Mirrahimi M, Beik J, Mirrahimi M, Alamzadeh Z, Teymouri S, Mahabadi VP, Eslahi N, Ebrahimi Tazehmahalleh F, Ghaznavi H, Shakeri-Zadeh A, Moustakis C. Triple combination of heat, drug and radiation using alginate hydrogel co-loaded with gold nanoparticles and cisplatin for locally synergistic cancer therapy. Int J Biol Macromol 2020; 158:617-626. [PMID: 32387354 DOI: 10.1016/j.ijbiomac.2020.04.272] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/23/2020] [Accepted: 04/30/2020] [Indexed: 01/14/2023]
Abstract
Although multimodal cancer therapy has shown superior antitumor efficacy in comparison to individual therapy due to the potential generation of synergistic interactions among the treatments, its clinical usage is highly hampered by systemic dose-limiting toxicities. Herein, we developed a multi-responsive nanocomplex constructed from alginate hydrogel co-loaded with cisplatin and gold nanoparticles (AuNPs) (abbreviated as ACA) to combine chemotherapy, radiotherapy (RT) and photothermal therapy. The nanocomplex markedly improved the efficiency of drug delivery where ACA resulted in noticeably higher tumor growth inhibition than free cisplatin. The tumor treated with ACA showed an increased heating rate upon 532 nm laser irradiation, indicating the photothermal conversion ability of the nanocomplex. While RT alone resulted in slight tumor growth inhibition, thermo-chemo therapy, chemoradiation therapy and thermo-radio therapy using ACA dramatically slowed down the rate of tumor growth. Upon 532 nm laser and 6 MV X-ray, the nanocomplex could enable a trimodal thermo-chemo-radio therapy that yielded complete tumor regression with no evidence of relapse during the 90-days follow up period. The results of this study demonstrated that the incorporation of AuNPs and cisplatin into alginate hydrogel network can effectively combine chemotherapy, RT and photothermal therapy to achieve a locally synergistic cancer therapy.
Collapse
Affiliation(s)
- Mehri Mirrahimi
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Jaber Beik
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mehraban Mirrahimi
- Biology Department, School of Science, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Alamzadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Samaneh Teymouri
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Vahid Pirhajati Mahabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Eslahi
- Endometriosis Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fatemeh Ebrahimi Tazehmahalleh
- Department of Radiooncology and Cyberknife Center, Faculty of Medicine, University Hospital Cologne, University Hospital Cologne, Cologne, Germany
| | - Habib Ghaznavi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences (ZaUMS), Zahedan, Iran.
| | - Ali Shakeri-Zadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Christos Moustakis
- Department of Radiation Oncology, University Hospital of Muenster, Muenster, Germany
| |
Collapse
|
32
|
Zhao P, Liu S, Wang L, Liu G, Cheng Y, Lin M, Sui K, Zhang H. Alginate mediated functional aggregation of gold nanoclusters for systemic photothermal therapy and efficient renal clearance. Carbohydr Polym 2020; 241:116344. [PMID: 32507204 DOI: 10.1016/j.carbpol.2020.116344] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/17/2020] [Accepted: 04/17/2020] [Indexed: 12/21/2022]
Abstract
For renal clearable nanoagents, it is challenging to delay the renal clearance to acquire efficient tumor accumulation. Herein, we report sodium alginate (SA) stabilized gold (Au) NCs. The Au NCs are of high biocompatibility and renal clearable. Contributed from the ligands of SA, the half-life (t1/2) of Au NCs is prolonged to ∼9.3 h, enhancing the tumor accumulation rate to 10.4 %ID/g. In tumor microenvironment (TME), the Au NCs are stimulated to functionally aggregate, which switches on the photothermal effect. Animal experiments prove that Au NCs aggregates are efficient photothermal therapy (PTT) agents for both local treatment of single tumors and systemic treatment of double-tumor models without causing noticeable side effects, confirming the biosecurity of Au NCs and systemic PTT. The switchable strategy of PTT may signify the establishment of a new systemic therapeutic methodology.
Collapse
Affiliation(s)
- Pin Zhao
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, College of Materials Science and Engineering, Institute of Marine Biobased Materials, Qingdao University, Qingdao 266071, PR China
| | - Shuwei Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Lu Wang
- Department of Oral Pathology, School and Hospital of Stomatology, Jilin University, Changchun 130021, PR China
| | - Guojian Liu
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, College of Materials Science and Engineering, Institute of Marine Biobased Materials, Qingdao University, Qingdao 266071, PR China
| | - Yanru Cheng
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, College of Materials Science and Engineering, Institute of Marine Biobased Materials, Qingdao University, Qingdao 266071, PR China
| | - Min Lin
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, College of Materials Science and Engineering, Institute of Marine Biobased Materials, Qingdao University, Qingdao 266071, PR China.
| | - Kunyan Sui
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Shandong Collaborative Innovation Center of Marine Biobased Fibers and Ecological Textiles, College of Materials Science and Engineering, Institute of Marine Biobased Materials, Qingdao University, Qingdao 266071, PR China.
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China.
| |
Collapse
|
33
|
Gold nanoparticles promote a multimodal synergistic cancer therapy strategy by co-delivery of thermo-chemo-radio therapy. Eur J Pharm Sci 2020; 145:105235. [DOI: 10.1016/j.ejps.2020.105235] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/16/2020] [Accepted: 01/22/2020] [Indexed: 12/21/2022]
|
34
|
Abdel-Ghany S, Mahfouz M, Ashraf N, Sabit H, Cevik E, El-Zawahri M. Gold nanoparticles induce G2/M cell cycle arrest and enhance the expression of E-cadherin in breast cancer cells. INORG NANO-MET CHEM 2020. [DOI: 10.1080/24701556.2020.1728553] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Mennatallah Mahfouz
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Nada Ashraf
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Hussein Sabit
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Emre Cevik
- Department of Genetics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Mokhtar El-Zawahri
- Department of Medical and Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
- Center for Research and Development, Misr University for Science and Technology, Giza, Egypt
| |
Collapse
|
35
|
Tang Q, Hu P, Peng H, Zhang N, Zheng Q, He Y. Near-Infrared Laser-Triggered, Self-Immolative Smart Polymersomes for in vivo Cancer Therapy. Int J Nanomedicine 2020; 15:137-149. [PMID: 32021170 PMCID: PMC6964533 DOI: 10.2147/ijn.s224502] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/14/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose Traditional chemotherapy is accompanied by significant side effects, which, in many aspects, limits its treatment efficacy and clinical applications. Herein, we report an oxidative responsive polymersome nanosystem mediated by near infrared (NIR) light which exhibited the combination effect of photodynamic therapy (PDT) and chemotherapy. Methods In our study, poly (propylene sulfide)20-bl-poly (ethylene glycol)12 (PPS20-b-PEG12) block copolymer was synthesized and employed to prepare the polymersome. The hydrophobic photosensitizer zinc phthalocyanine (ZnPc) was loaded in the shell and the hydrophilic doxorubicin hydrochloride (DOX·HCl) in the inner aqueous space of the polymersome. Results Under the irradiation of 660 nm NIR light, singlet oxygen 1O2 molecules were generated from ZnPc to oxidize the neighbouring sulfur atoms on the PPS block which eventually ruptured the intact structure of polymersomes, leading to the release of encapsulated DOX·HCl. The released DOX and the 1O2 could achieve a combination effect for cancer therapy if the laser activation and drug release occur at the tumoral sites. In vitro studies confirmed the generation of singlet oxygen and DOX release by NIR irradiation. In vivo studies showed that such a combined PDT-chemotherapy nanosystem could accumulate in A375 tumors efficiently, thus leading to significant inhibition on tumor growth as compared to PDT (PZ group) or chemotherapy alone (DOX group). Conclusion In summary, this oxidation-sensitive nanosystem showed excellent anti-tumor effects by synergistic chemophotodynamic therapy, indicating that this novel drug delivery strategy could potentially provide a new means for cancer treatments in clinic.
Collapse
Affiliation(s)
- Qing Tang
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Ping Hu
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China.,School of Pharmacy, Jinan University, Guangzhou 510632, People's Republic of China
| | - Haibo Peng
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Ning Zhang
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Qiang Zheng
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| | - Yun He
- School of Pharmaceutical Sciences and Chongqing Key Laboratory of Natural Drug Research, Chongqing University, Chongqing 401331, People's Republic of China
| |
Collapse
|
36
|
Mérida F, Rinaldi C, Juan EJ, Torres-Lugo M. In vitro Ultrasonic Potentiation of 2-Phenylethynesulfonamide/Magnetic Fluid Hyperthermia Combination Treatments for Ovarian Cancer. Int J Nanomedicine 2020; 15:419-432. [PMID: 32021188 PMCID: PMC6982443 DOI: 10.2147/ijn.s217870] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/25/2019] [Indexed: 01/15/2023] Open
Abstract
Background Magnetic Fluid Hyperthermia (MFH) is a promising adjuvant for chemotherapy, potentiating the action of anticancer agents. However, drug delivery to cancer cells must be optimized to improve the overall therapeutic effect of drug/MFH combination treatments. Purpose The aim of this work was to demonstrate the potentiation of 2-phenylethynesulfonamide (PES) at various combination treatments with MFH, using low-intensity ultrasound as an intracellular delivery enhancer. Methods The effect of ultrasound (US), MFH, and PES was first evaluated individually and then as combination treatments. Definity® microbubbles and polyethylene glycol (PEG)-coated iron oxide nanoparticles were used to induce cell sonoporation and MFH, respectively. Assessment of cell membrane permeabilization was evaluated via fluorescence microscopy, iron uptake by cells was quantified by UV-Vis spectroscopy, and cell viability was determined using automatic cell counting. Results Notable reductions in cancer cell viability were observed when ultrasound was incorporated. For example, the treatment US+PES reduced cell viability by 37% compared to the non-toxic effect of the drug. Similarly, the treatment US+MFH using mild hyperthermia (41°C), reduced cell viability by an additional 18% when compared to the effect of MH alone. Significant improvements were observed for the combination of US+PES+MFH with cell viability reduced by an additional 26% compared to the PES+MFH group. The improved cytotoxicity was attributed to enhanced drug/nanoparticle intracellular delivery, with iron uptake values nearly twice those achieved without ultrasound. Various treatment schedules were examined, and all of them showed substantial cell death, indicating that the time elapsed between sonoporation and magnetic field exposure was not significant. Conclusion Superior cancer cell-killing patterns took place when ultrasound was incorporated thus demonstrating the in vitro ultrasonic potentiation of PES and mild MFH. This work demonstrated that ultrasound is a promising non-invasive enhancer of PES/MFH combination treatments, aiming to establish a sono-thermo-chemotherapy in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Fernando Mérida
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| | - Carlos Rinaldi
- Department of Chemical Engineering, University of Florida, Gainesville, FL, USA.,J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Eduardo J Juan
- Department of Electrical and Computer Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| | - Madeline Torres-Lugo
- Department of Chemical Engineering, University of Puerto Rico-Mayagüez, Mayagüez, Puerto Rico
| |
Collapse
|
37
|
Song C, Tang C, Xu W, Ran J, Wei Z, Wang Y, Zou H, Cheng W, Cai Y, Han W. Hypoxia-Targeting Multifunctional Nanoparticles for Sensitized Chemotherapy and Phototherapy in Head and Neck Squamous Cell Carcinoma. Int J Nanomedicine 2020; 15:347-361. [PMID: 32021184 PMCID: PMC6980849 DOI: 10.2147/ijn.s233294] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose Chemotherapy in head and neck squamous cell carcinoma (HNSCC) has many systemic side effects, as well as hypoxia-induced chemoresistance. To reduce side effects and enhance chemosensitivity are urgently needed. Methods We synthesized a drug delivery system (named CECMa NPs) based on cisplatin (CDDP) and metformin (chemotherapeutic sensitizer), of which chlorin e6 (Ce6) and polyethylene glycol diamine (PEG) were synthesized as the shell, an anti-LDLR antibody (which can target to hypoxic tumor cells) was modified on the surface to achieve tumor targeting. Results The NPs possessed a great synergistic effect of chemotherapy and phototherapy. After laser stimulation, both CDDP and metformin can be released in situ to achieve anti-tumor effects. Meanwhile, PDT and PTT triggered by a laser have anticancer effects. Furthermore, compared with free cisplatin, CECMa exhibits less systemic toxicity with laser irradiation in the xenograft mouse tumor model. Conclusion CECMa effectively destroyed the tumors via hypoxia targeting multimodal therapy both in vitro and in vivo, thereby providing a novel strategy for targeting head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
- Chuanhui Song
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Chuanchao Tang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Wenguang Xu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Jianchuan Ran
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Zheng Wei
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China.,Pediatric Dentistry, Nanjing Stomatology Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Yufeng Wang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Huihui Zou
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Wei Cheng
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Yu Cai
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| | - Wei Han
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, People's Republic of China
| |
Collapse
|
38
|
Guo X, Mei J, Zhang C. Development of Drug Dual-Carriers Delivery System with Mitochondria-Targeted and pH/Heat Responsive Capacity for Synergistic Photothermal-Chemotherapy of Ovarian Cancer. Int J Nanomedicine 2020; 15:301-313. [PMID: 32021181 PMCID: PMC6970626 DOI: 10.2147/ijn.s226517] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/28/2019] [Indexed: 01/04/2023] Open
Abstract
PURPOSE Multifunctional drug delivery systems (DDS) are emerging as a new strategy to highly treat malignant tumors. The aim of this study is to develop a drug dual-carriers delivery system (DDDS) using the natural protein ferritin (FRT) and a nanoscale graphene oxide (NGO) as dual-carriers. METHODS The FRT is a pH-sensitive hollow cage protein with disassembly and reassembly properties and the NGO has a large surface area and a photothermal effect by which it can load and release drugs under near-infrared irradiation (NIR). Due to these unique features, the NGO loaded the anticancer drug resveratrol (RSV) and the conjugated mitochondrion targeted molecule IR780 as IR780-NGO-RSV (INR), the first drug delivery platform. Next, the INR was capsulated by FRT to form the DDDS INR@FRT which was applied for synergistic photothermal-chemotherapy of ovarian cancer. RESULTS Through a series of characterizations, INR@FRT showed a uniform nanosphere structure and remarkable stability in physiological condition. Heat/pH 5.0 was confirmed to trigger RSV release from the INR@FRT. After taken up by cells, INR@FRT located to the lysosomes where the acidic environment triggered INR release. INR targeted the mitochondrion and released RSV to directly react with organelles, which in turn decreased the mitochondrion membrane potential and caused cell apoptosis. In-vivo experiments showed that INR@FRT combined with NIR irradiation displayed remarkable tumor suppression with a high survival rate after 60 days of treatment. Finally, the biocompatibility of INR@FRT was demonstrated in vitro and in vivo. CONCLUSION These results highlight the immense potential of INR@FRT as a type of DDDS for the treatment of tumors.
Collapse
Affiliation(s)
- Xiaoxia Guo
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu610041, Sichuan, People’s Republic of China
| | - Jie Mei
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu610041, Sichuan, People’s Republic of China
| | - Chunping Zhang
- The Center of Clinical Laboratory, Sichuan Great Master Diagnostics Co. Ltd, Chengdu611731, Sichuan, People’s Republic of China
| |
Collapse
|
39
|
Cai Q, Wang X, Wang S, Jin L, Ding J, Zhou D, Ma F. Gallbladder Cancer Progression Is Reversed by Nanomaterial-Induced Photothermal Therapy in Combination with Chemotherapy and Autophagy Inhibition. Int J Nanomedicine 2020; 15:253-262. [PMID: 32021178 PMCID: PMC6970248 DOI: 10.2147/ijn.s231289] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction Gallbladder cancer (GBC) is the most common malignancy in biliary tract with extremely poor prognosis. Photothermal therapy (PTT) shows great promises for tumor therapy, which causes tumor cell death via selectively directed heating released by nanoparticles under the near-infrared irradiation. Through degrading damaged organelles and misfolded proteins in autophagosomes, autophagy plays a vital role in maintaining the intracellular homeostasis. The present study attempted to combine chemotherapy and autophagy blocking with PTT. Materials and Methods We purchased multi-walled carbon nanotubes from Nanostructured and Amorphous Materials and performed PTT using an 808-nm diode laser. The cytotoxic effects of PTT and chemotherapy in vitro were assessed by cell viability analysis. The effects of PTT and chemotherapy on autophagy in vitro were assessed by GFP-LC3 and Western blot. And these results were confirmed by in vivo experiment. Results Both PTT and chemotherapy could trigger cytoprotective autophagy to tolerate the cellular stresses and prolong the survival of GBC cell; therefore, the blocking of autophagy could enhance the efficacy of PTT and chemotherapy in GBC treatment in vitro and in vivo. Conclusion Chemotherapeutic drug doxorubicin and autophagy inhibitor chloroquine could enhance the efficacy of nanoparticle-mediated hyperthermia in GBC.
Collapse
Affiliation(s)
- Qiang Cai
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China.,Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Xinjing Wang
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Longyang Jin
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Jun Ding
- Department of Biliary and Pancreatic Surgery, Shanghai Shuguang Hospital Affiliated with Shanghai University of T.C.M., Shanghai 201203, People's Republic of China
| | - Di Zhou
- Department of General Surgery, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200092, People's Republic of China
| | - Fei Ma
- Department of Oncology, Xinhua Hospital, Shanghai JiaoTong University School of Medicine, Shanghai Institute for Pediatric Research, Shanghai 200092, People's Republic of China
| |
Collapse
|
40
|
Sheervalilou R, Shahraki O, Hasanifard L, Shirvaliloo M, Mehranfar S, Lotfi H, Pilehvar-Soltanahmadi Y, Bahmanpour Z, Zadeh SS, Nazarlou Z, Kangarlou H, Ghaznavi H, Zarghami N. Electrochemical Nano-biosensors as Novel Approach for the Detection of Lung Cancer-related MicroRNAs. Curr Mol Med 2019; 20:13-35. [DOI: 10.2174/1566524019666191001114941] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/22/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
In both men and women around the world, lung cancer accounts as the
principal cause of cancer-related death after breast cancer. Therefore, early detection of
the disease is a cardinal step in improving prognosis and survival of patients. Today, the
newly-defined microRNAs regulate about 30 to 60 percent of the gene expression.
Changes in microRNA Profiles are linked to numerous health conditions, making them
sophisticated biomarkers for timely, if not early, detection of cancer. Though evaluation
of microRNAs in real samples has proved to be rather challenging, which is largely
attributable to the unique characteristics of these molecules. Short length, sequence
similarity, and low concentration stand among the factors that define microRNAs.
Recently, diagnostic technologies with a focus on wide-scale point of care have recently
garnered attention as great candidates for early diagnosis of cancer. Electrochemical
nano-biosensors have recently garnered much attention as a molecular method,
showing great potential in terms of sensitivity, specificity and reproducibility, and last but
not least, adaptability to point-of-care testing. Application of nanoscale materials in
electrochemical devices as promising as it is, brings multiplexing potential for conducting
simultaneous evaluations on multiple cancer biomarkers. Thanks to their enthralling
properties, these materials can be used to improve the efficiency of cancer diagnostics,
offer more accurate predictions of prognosis, and monitor response to therapy in a more
efficacious way. This article presents a concise overview of recent advances in the
expeditiously evolving area of electrochemical biosensors for microRNA detection in
lung cancer.
Collapse
Affiliation(s)
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Leili Hasanifard
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Shirvaliloo
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Mehranfar
- Department of Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hajie Lotfi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Younes Pilehvar-Soltanahmadi
- Cellular and Molecular Research Center, Research Institute for Cellular and Molecular Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zahra Bahmanpour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadaf Sarraf Zadeh
- Neurosciences Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ziba Nazarlou
- Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey
| | - Haleh Kangarlou
- Department of Physics, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Nosratollah Zarghami
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Gao J, Liu J, Xie F, Lu Y, Yin C, Shen X. Co-Delivery of Docetaxel and Salinomycin to Target Both Breast Cancer Cells and Stem Cells by PLGA/TPGS Nanoparticles. Int J Nanomedicine 2019; 14:9199-9216. [PMID: 32063706 PMCID: PMC6884979 DOI: 10.2147/ijn.s230376] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/13/2019] [Indexed: 01/27/2023] Open
Abstract
PURPOSE Conventional chemotherapy is hampered by the presence of breast cancer stem cells (BCSCs). It is crucial to eradicating both the bulky breast cancer cells and BCSCs, using a combination of conventional chemotherapy and anti-CSCs drugs. However, the synergistic ratio of drug combinations cannot be easily maintained in vivo. In our previous studies, we demonstrated that the simultaneous delivery of two drugs via nanoliposomes could maintain the synergistic drug ratio for 12 h in vivo. However, nanoliposomes have the disadvantage of quick drug release, which makes it difficult to maintain the synergistic drug ratio for a long time. Herein, we developed a co-delivery system for docetaxel (DTX)-a first-line chemotherapy drug for breast cancer-and salinomycin (SAL)-an anti-BCSCs drug-in rigid nanoparticles constituted of polylactide-co-glycolide/D-alpha-tocopherol polyethylene glycol 1000 succinate (PLGA/TPGS). METHODS Nanoparticles loaded with SAL and DTX at the optimized ratio (NSD) were prepared by the nanoprecipitation method. The characterization, cellular uptake, and cytotoxicity of nanoparticles were investigated in vitro, and the pharmacokinetics, tissue distribution, antitumor and anti-CSCs activity of nanoparticles were evaluated in vivo. RESULTS We demonstrated that a SAL/DTX molar ratio of 1:1 was synergistic in MCF-7 cells and MCF-7-MS. Moreover, the enhanced internalization of nanoparticles was observed in MCF-7 cells and MCF-7-MS. Furthermore, the cytotoxicity of NSD against both MCF-7 cells and MCF-7-MS was stronger than the cytotoxicity of any single treatment in vitro. Significantly, NSD could prolong the circulation time and maintain the synergistic ratio of SAL to DTX in vivo for 24 h, thus exhibiting superior tumor targeting and anti-tumor activity compared to other treatments. CONCLUSION Co-encapsulation of SAL and DTX in PLGA/TPGS nanoparticles could maintain the synergistic ratio of drugs in vivo in a better manner; thus, providing a promising strategy for synergistic inhibition of breast cancer.
Collapse
Affiliation(s)
- Jie Gao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Scientific Research Center, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| | - Junjie Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai, People’s Republic of China
| | - Fangyuan Xie
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, People’s Republic of China
| | - Ying Lu
- Department of Pharmaceutical Sciences, Second Military Medical University, Shanghai, People’s Republic of China
| | - Chuan Yin
- Department of Gastroenterology, Changzheng Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Zhejiang, People’s Republic of China
| |
Collapse
|
42
|
Zhang L, Yang P, Guo R, Sun J, Xie R, Yang W. Multifunctional Mesoporous Polydopamine With Hydrophobic Paclitaxel For Photoacoustic Imaging-Guided Chemo-Photothermal Synergistic Therapy. Int J Nanomedicine 2019; 14:8647-8663. [PMID: 31806962 PMCID: PMC6839591 DOI: 10.2147/ijn.s218632] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 10/05/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chemo-photothermal therapy has attracted intensive attention because of its low side effects and better therapeutic efficiency. Although many photothermal agents have been loaded with chemotherapeutic drugs for chemo-photothermal therapy, their applications are limited by complex synthetic protocols and long-term safety. Therefore, there is significant clinical value in the development of a simple system of biocompatible and biodegradable photothermal nanomaterials with high payloads of chemotherapeutic drugs for chemo-photothermal synergistic therapy. MATERIALS AND METHODS In this study, PEG-modified polydopamine nanoparticles with mesoporous structure (MPDA-PEG) were successfully obtained by an emulsion-induced interface assembly strategy. Subsequently, paclitaxel (PTX) dissolved in acetone was loaded into the mesoporous channels of MPDA-PEG nanoparticles by solution absorption method. A PTX-loaded MPDA-PEG (MPDA-PEG-PTX) nanoplatform for combination of photothermal therapy (PTT) and chemotherapy was developed. RESULTS The synthesized MPDA-PEG nanoparticles had a great photothermal effect under near-infrared (NIR) laser irradiation and exhibited an enhanced photothermal effect with the increase of particle size. Meanwhile, MPDA-PEG nanoparticles also had a high payload of PTX, and the PTX release could be greatly accelerated by elevated temperature from photothermal effect. In MTT cytotoxicity assay, A549 cells incubated with MPDA-PEG-PTX under NIR laser irradiation (PTT + chemotherapy group) exhibited better therapeutic effect than single chemotherapy (MPDA-PEG-PTX group) and PTT (MPDA-PEG + Laser group). The synergistic therapeutic effect of MPDA-PEG-PTX with NIR laser irradiation in vivo was further investigated under the guidance of photoacoustic imaging (PAI), tumors of nude mice treated with MPDA-PEG-PTX with NIR laser irradiation were completely eliminated with minimal side effect. CONCLUSION The MPDA-PEG-PTX nanoplatform is a simple and effective platform which can completely inhibit tumor growth with minimal side effects under NIR irradiation, and it exhibits better therapeutic effect than single chemotherapy and PTT.
Collapse
Affiliation(s)
- Liren Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200433, People’s Republic of China
| | - Peng Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200433, People’s Republic of China
| | - Ranran Guo
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200433, People’s Republic of China
| | - Jiaxin Sun
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200433, People’s Republic of China
| | - Ruihong Xie
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200433, People’s Republic of China
| | - Wuli Yang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200433, People’s Republic of China
| |
Collapse
|
43
|
Asadi M, Beik J, Hashemian R, Laurent S, Farashahi A, Mobini M, Ghaznavi H, Shakeri-Zadeh A. MRI-based numerical modeling strategy for simulation and treatment planning of nanoparticle-assisted photothermal therapy. Phys Med 2019; 66:124-132. [DOI: 10.1016/j.ejmp.2019.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
|
44
|
Simulation-guided photothermal therapy using MRI-traceable iron oxide-gold nanoparticle. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 199:111599. [DOI: 10.1016/j.jphotobiol.2019.111599] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 11/21/2022]
|
45
|
Jie S, Guo X, Ouyang Z. Tumor ablation using novel photothermal Na xWO 3 nanoparticles against breast cancer osteolytic bone metastasis. Int J Nanomedicine 2019; 14:7353-7362. [PMID: 31571856 PMCID: PMC6750009 DOI: 10.2147/ijn.s217974] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Backgrounds Profiting from the development of nanomaterials, photothermal therapy (PTT) has been discovered as efficient tumor ablation strategy for breast cancer. Materials and methods Novel oxygen vacancy-rich tungsten bronze nanoparticles (NaxWO3) were synthesized through a simple pyrogenic decomposition process. TEM, XRD, UV-vis-NIR, photothermal conversion ability, and photothermal stability were performed. The viabilities of 293T and 4T1 cells after treating with 200 μg/mL NaxWO3 nanoparticles for 24 or 48 hrs were both above 80%, which proved the good biosafety and cytotoxicity of NaxWO3 in vitro. Two in vivo breast cancer models, namely percutaneous and intratibial 4T1 models were established and NaxWO3 (20 mg/kg) with power intensity of 1.5 W/cm2 980 nm laser photothermal treatment was used in vivo. Results We successfully synthesized ~150 nm NaxWO3 nanoparticles with desirable PTT effects, as evidenced by the temperature increase from 25.8°C to 41.8°C in 5 mins under the irradiation of 980 nm laser (1 mg/mL). Also, cellular compatibility of NaxWO3 nanoparticles was found upon physiologic 293T cells, in contrast with significant cytotoxicity against breast cancer 4T1 cell in vitro dose-dependently. Besides, two in vivo breast cancer models showed the decent tumor ablation ability of NaxWO3 nanoparticles, demonstrating percutaneous 4T1 tumor elimination without recurrence during 2 weeks observation as well as intratibial breast cancer inhibition with decreased bone destruction and tumor volume after NaxWO3+PTT in vivo. Conclusion For the first time, we developed a novel oxygen vacancy-rich tungsten bronze nanoparticles (NaxWO3) through a simple pyrogenic decomposition process for PTT. Both in vitro and in vivo experiments showed the good PTT ability and tumor ablation effects of synthesized NaxWO3 nanoparticles against breast cancer osteolytic bone metastasis. Additionally, our oxygen-deficient NaxWO3 nanoparticles will expand the research horizons of PTT nanomaterials.
Collapse
Affiliation(s)
- Shuo Jie
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Xiaoning Guo
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| | - Zhengxiao Ouyang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, People's Republic of China
| |
Collapse
|
46
|
Ardakani AA, Ghader A, Asgari H, Keshavarz M, Tazehmahalleh FE, Majles Ara MH, Malekzadeh M, Ghaznavi H, Shakeri-Zadeh A. The capability of nonlinear optical characteristics as a predictor for cellular uptake of nanoparticles and cell damage. Photodiagnosis Photodyn Ther 2019; 27:442-448. [PMID: 31362112 DOI: 10.1016/j.pdpdt.2019.07.023] [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: 04/15/2019] [Revised: 07/21/2019] [Accepted: 07/26/2019] [Indexed: 11/19/2022]
Abstract
Current methods for determining the cellular effects of a treatment modality need expensive materials and much time to provide a researcher with results. The aim of this study was to evaluate the potential of nonlinear optical characteristics of cancer cells using Z-scan technique to monitor the level of cellular uptake and cell damage caused by a nanotechnology based treatment modality. Two nanocomplexes were synthesized and characterized. The first one was made of alginate hydrogel co-loaded with cisplatin and gold nanoparticles (AuNPs) named as ACA nanocomplex. The second one, named as AA nanocomplex, was the same as ACA, but without cisplatin and this AA nanocomplex was considered as the control for ACA. Different groups of CT26 mouse colon cancer cell line received various treatments of cisplatin, ACA, and AA nanocomplexes and then the samples were prepared for Z-scan studies. The MTT assay was used to evaluate the cytotoxicity induced by different treatment modalities. Transmission electron microscopy (TEM) and inductively coupled plasma-mass spectrometry (ICP-MS) were used for qualitative and quantitative assessments of the level of AuNPs cellular uptake. The trend of nonlinear optical properties changes for treated cells was in agreement with MTT, TEM and ICP-MS results. Z-scan technique was able to successfully indicate the occurrence of cell damage. It was also capable to determine the intensity of cell damage induced by ACA nanocomplex in comparison to free cisplatin. Furthermore, Z-scan results showed that it was able to discriminate the differences of optical properties of the cells incubated with ACA nanocomplex for various incubation times. Nonlinear optical characteristics of a cell may be considered as a reliable indicator to predict the level of cellular effects induced by a nanotechnology based treatment modality. The protocol suggested in this article does not waste materials, not take much time to provide the results, and it is inexpensive technique.
Collapse
Affiliation(s)
- Ali Abbasian Ardakani
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Alireza Ghader
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Hamid Asgari
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Marzieh Keshavarz
- Department of Medical Physics, School of Medicine, Shiraz University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Hosein Majles Ara
- Department of Physics, Biophotonics Lab, Applied Science Research Center (ASRC), Kharazmi University, Karaj, Iran
| | - Malekeh Malekzadeh
- 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
- Finetech in Medicine Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
47
|
Xu Z, Hu C, Chen S, Zhang C, Yu J, Wang X, Lv H, Cheng X. Apatinib enhances chemosensitivity of gastric cancer to paclitaxel and 5-fluorouracil. Cancer Manag Res 2019; 11:4905-4915. [PMID: 31213909 PMCID: PMC6549793 DOI: 10.2147/cmar.s196372] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/06/2019] [Indexed: 12/15/2022] Open
Abstract
Background and aim: Paclitaxel (PTX) plus 5-fluorouracil (5-Fu) has become the standard chemotherapy for advanced gastric cancer (GC). Apatinib, a small-molecule tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor-2, improves outcomes in GC patients as a third-line treatment. However, its impact on the chemosensitivity of GC remains to be determined. Hence, we aimed to assess the efficacy and safety of apatinib combined with chemotherapy in vivo and in vitro. Methods: The MGC803 cell viability was determined by Cell Counting Kit-8 assay, and the interactions between apatinib and conventional cytotoxic agents revealed by combination index values were calculated using Calcusyn 2.0 software. We also used a zebrafish embryo xenograft model to validate the synergistic interactions. Furthermore, 4 patients with late-stage GC were enrolled to explore the efficacy and safety of PTX/Tegafur Gimeracil Oteracil Potassium (S1) (PS) chemotherapy plus apatinib in conversion surgery. Results: Apatinib showed synergistic interactions with both PTX and 5-Fu in vivo. The zebrafish embryo xenograft model also demonstrated that apatinib significantly enhanced the antitumor activity of PTX and 5-Fu. Apatinib plus PS chemotherapy was well tolerated before surgery. Objective response to preoperative SPA treatment was achieved in all 4 patients. No postoperative bleeding events or wound-healing complications were observed. No postperative morbidity occurred and no morbidity was encountered. Pathological examination showed that all patients had grade Ib pathological response. Conclusion: The experimental data suggested that apatinib improves the efficacy of PTX and 5-Fu both in vitro and in vivo. Clinical evidence showed that a combination of PS chemotherapy with apatinib may be an efficient and acceptable safety treatment for late-stage GC, especially in conversion surgery.
Collapse
Affiliation(s)
- Zhiyuan Xu
- Department of Abdominal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Can Hu
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Shangqi Chen
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Chunli Zhang
- Department of Pathology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianfa Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaofeng Wang
- First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Hang Lv
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Diagnosis and Treatment of Digestive System Tumor, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Xiangdong Cheng
- Department of Abdominal Surgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
48
|
Li XD, Wang Z, Wang XR, Shao D, Zhang X, Li L, Ge MF, Chang ZM, Dong WF. Berberine-loaded Janus gold mesoporous silica nanocarriers for chemo/radio/photothermal therapy of liver cancer and radiation-induced injury inhibition. Int J Nanomedicine 2019; 14:3967-3982. [PMID: 31239666 PMCID: PMC6554520 DOI: 10.2147/ijn.s206044] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 04/13/2019] [Indexed: 12/21/2022] Open
Abstract
Background: The combination of chemotherapy with radiotherapy serves as a common therapeutic strategy in clinics. However, it is unsatisfactory due to its poor therapeutic efficiency and severe side-effects originating from chemotherapy-exerted systemic toxicity as well as radiation-induced injury. Purpose: Hence, Berberine (Ber), an isoquinolin alkaloid with low toxicity and protective effects against radiotherapy, was used as a novel chemotherapeutic agent for chemo-radiotherapy of liver cancer. Patients and methods: We preloaded Ber into folic acid targeting Janus gold mesoporous silica nanocarriers (FA-JGMSNs) for overcoming the poor bioavailability of Ber. Furthermore, FA-JGMSNs were not only employed as radiosensitizers for expanding radiotherapeutic effect, but also used as photothermal agents for supplementing chemo-radiotherapeutic effect by local photothermal therapy. Results: In vitro and in vivo experiemtal results demonstrated the highly efficient anti-tumor effect, good biosafety as well as the effective protection of normal tissue of this nanoplatform. Conclusion: Based on its superb performance, we believe our work provided a feasible strategy for triple-therapies of liver cancer.
Collapse
Affiliation(s)
- Xiao-Dong Li
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing100049, People’s Republic of China
- Department of Echocardiography, The First Hospital of Jilin University, Changchun130021, People’s Republic of China
| | - Zheng Wang
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing100049, People’s Republic of China
| | - Xin-Rui Wang
- Department of Hepatology, The First Hospital of Jilin University, Changchun130021, People’s Republic of China
| | - Dan Shao
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, People’s Republic of China
| | - Xi Zhang
- Department of Rheumatology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou510630, People’s Republic of China
| | - Li Li
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, People’s Republic of China
| | - Ming-Feng Ge
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, People’s Republic of China
| | - Zhi-Min Chang
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, People’s Republic of China
| | - Wen-Fei Dong
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou215163, People’s Republic of China
| |
Collapse
|
49
|
Zhang Y, Wang M, Xu X, Liu Y, Xiao C. Matrine promotes apoptosis in SW480 colorectal cancer cells via elevating MIEF1-related mitochondrial division in a manner dependent on LATS2-Hippo pathway. J Cell Physiol 2019; 234:22731-22741. [PMID: 31119752 DOI: 10.1002/jcp.28838] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/26/2019] [Accepted: 04/30/2019] [Indexed: 12/16/2022]
Abstract
Matrine, an alkaloid compound isolated from Sophora flavescens Ait, has been shown to exert cancer-killing actions in a variety of tumors; however, its anticancer mechanism in colorectal cancer (CRC) is not clear. The goal of our study was to characterize the anticancer effects and molecular mechanisms of matrine in SW480 CRC cells in vitro. Matrine treatment reduced mitochondrial metabolic function and ATP levels, repressed mitochondrial membrane potential, evoked mitochondrial reactive oxygen species accumulation, and promoted cyt-c-related mitochondrial apoptosis activation. In addition, we found that matrine treatment activated mitochondrial fission through upregulating mitochondrial elongation factor 1 (MIEF1); silencing of MIEF1 prevented matrine-mediated mitochondrial damage and reversed the decrease in SW480 cell viability. Moreover, matrine treatment affected MIEF1 expression via the large tumor suppressor-2 (LATS2)-Hippo axis, and LATS2 deficiency suppressed the anticancer actions exerted by matrine on SW480 cancer cells. In summary, we show for the first time that matrine inhibits SW480 cell survival by activating MIEF1-related mitochondrial division via the LATS2-Hippo pathway. These findings explain the anticancer mechanisms of matrine in CRC and also identify the LATS2-MIEF1 signaling pathway as an effective target for the treatment of CRC.
Collapse
Affiliation(s)
- Yawei Zhang
- Department of General Surgery, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Meiping Wang
- Department of General Surgery, Fuzhou General Hospital (Dongfang Hospital), Fuzhou, Fujian, China
| | - Xianfeng Xu
- Department of Critical Care Medicine, Changle People's Hospital, Fuzhou, Fujian, China
| | - Yonghong Liu
- Department of General Surgery, First People's Hospital of Yuhang District, Hangzhou, China
| | - Chunhong Xiao
- Department of General Surgery, Fuzhou General Hospital (Dongfang Hospital), Fuzhou, Fujian, China
| |
Collapse
|
50
|
Mirrahimi M, Abed Z, Beik J, Shiri I, Shiralizadeh Dezfuli A, Mahabadi VP, Kamran Kamrava S, Ghaznavi H, Shakeri-Zadeh A. A thermo-responsive alginate nanogel platform co-loaded with gold nanoparticles and cisplatin for combined cancer chemo-photothermal therapy. Pharmacol Res 2019; 143:178-185. [DOI: 10.1016/j.phrs.2019.01.005] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/16/2018] [Accepted: 01/02/2019] [Indexed: 11/26/2022]
|