1
|
Yang C, Liao X, Zhou K, Yao Y, He X, Zhong W, Zheng D, Yang Y, Li M, Zhou M, Zhou Y, Li L, Bai Y, Shi K, Qian Z. Multifunctional nanoparticles and collagenase dual loaded thermosensitive hydrogel system for enhanced tumor-penetration, reversed immune suppression and photodynamic-immunotherapy. Bioact Mater 2025; 48:1-17. [PMID: 40028237 PMCID: PMC11870144 DOI: 10.1016/j.bioactmat.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
Breast cancer is the most prevalent and lethal malignancy among females, with a critical need for safer and less invasive treatments. Photodynamic therapy (PDT) can effectively eliminate tumor cells with minimal side effects. Furthermore, the combination of PDT and immunotherapy using nanoparticles has shown promise in treating both primary and distant metastatic tumor cells. Therefore, this study proposes applying the PDT-immunotherapy combination to breast cancer treatment. However, the low immunogenicity characteristic of "cold" tumors in part of breast cancer significantly diminishes therapeutic efficacy. To address this challenge, here, a nano-gel system (designated as HCSC-gel) is constructed, which co-delivers a mitochondria-targeted photosensitizer and a STING agonist, capable of robustly activating "cold" tumor immunity. This system is further enhanced by collagenase (CN) to improve therapeutic outcomes. Upon injection into the primary tumor site, HCSC-gel rapidly forms a gel matrix, releasing CN to degrade the tumor extracellular matrix and facilitate the penetration of photosensitizers, STING agonists, and oxygen into the tumor tissue. Under laser irradiation, PDT and STING-mediated immune responses are activated, reversing the low immunogenicity of breast cancer and effectively treating both primary and metastatic lesions. This HCSC-gel nano hydrogel delivery platform is anticipated to provide novel insights for the clinical management of breast cancer and other low immunogenic "cold" tumors, offering significant benefits to patients.
Collapse
Affiliation(s)
- Chengli Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Xukun Liao
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Kai Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Yongchao Yao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Xinlong He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Wen Zhong
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Dan Zheng
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yan Yang
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Ming Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Meng Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yadi Zhou
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Lin Li
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yang Bai
- Department of Pharmacy, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, And Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610041, China
| |
Collapse
|
2
|
Mendes M, António M, Daniel-da-Silva AL, Sereno J, Oliveira R, Arnaut LG, Gomes C, Ramos ML, Castelo-Branco M, Sousa J, Pais A, Vitorino C. A switch-on chemo-photothermal nanotherapy impairs glioblastoma. MATERIALS HORIZONS 2025. [PMID: 40202681 DOI: 10.1039/d5mh00351b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
Judiciously combined modality approaches have proved highly effective for treating most forms of cancer, including glioblastoma. This study introduces a hybrid nanoparticle-based treatment designed to induce a synergistic effect. It employs repurposed celecoxib-loaded hybrid nanoparticles (HNPs) that are thermally activated by near-infrared laser irradiation to damage glioblastoma cells. The HNPs are constructed by covalently binding organic (ultra-small nanostructured lipid carriers, usNLCs) and inorganic nanoparticles (gold nanorods, AuNRs, with photothermal therapy capability), using c(RGDfK) that serves the dual purpose of a biolinker and a tumor-targeting peptide. The HNPs are further functionalized with transferrin (Tf) as a blood-brain barrier ligand denoted as HNPsTf. Our comprehensive in vitro and in vivo studies have unveiled the remarkable capability of HNPsTf to safely and specifically increase blood-brain barrier permeability through transferrin receptor interactions, facilitating precise nanoparticle accumulation in the tumor region within orthotopic tumor-bearing mice. Furthermore, the orchestrated combination of chemo- and photothermal therapy has exhibited a substantial therapeutic impact on glioblastoma, showcasing a noteworthy 78% inhibition in tumor volume growth and an impressive 98% delay in tumor growth. Notably, this treatment approach has resulted in prolonged survival rates among tumor-bearing mice, accompanied by a favorable side effect profile. Overall, our findings unequivocally demonstrate that celecoxib-loaded HNPsTf offer a game-changing, chemo-photothermal combination, unleashing a synergistic effect that significantly enhances both brain drug delivery and the efficacy of anti-glioblastoma treatments.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Maria António
- CICECO-Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana L Daniel-da-Silva
- CICECO-Aveiro Institute of Materials, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - José Sereno
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Rui Oliveira
- Coimbra Institute for Clinical and Biomedical Research (iCBR)/Center of Environmental Genetics of Oncobiology (CIMAGO), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Germano de Sousa Pathological Anatomy Center, Quinta de Voimarães, Rua de S. Teotónio, 3000-377 Coimbra, Portugal
| | - Luís G Arnaut
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Célia Gomes
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
- Germano de Sousa Pathological Anatomy Center, Quinta de Voimarães, Rua de S. Teotónio, 3000-377 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Luísa Ramos
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548 Coimbra, Portugal
- Institute of Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, 3000-548 Coimbra, Portugal
| | - João Sousa
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Alberto Pais
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, 3000-535 Coimbra, Portugal
| |
Collapse
|
3
|
Guo G, Xu W, Fu J, Ma S, Huang K, Wei Y, Yang Y, Lan X, He X. A novel polysaccharide from Macadamia peel: Extraction, purification, structural characterization and antioxidant activity. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2025; 105:2681-2696. [PMID: 39579006 DOI: 10.1002/jsfa.14040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Macadamia peels are the main by-product of postharvest treatment of the whole fresh fruit weight, they contain various bioactive substances, such as polysaccharides, phenols, flavonoids, phenolic acids, saponins, and other nutritional and functional components, that are known to have anti-tumor and anti-oxidation functions. RESULTS Two purified polysaccharide fractions were obtained (MPP-1 and MPP-2) by extracting with ultrasonic-microwave-aided water extraction and purifying by with DEAE-52 and Sephadex G-50 columns, and then characterized using Fourier-transform infrared spectroscopy, scanning electron microscopy, atomic force microscopy and NMR, and the antioxidant activity was also investigated. The results indicated that MPP-1 and MPP-2 were mainly glucose, the molecular weight was 8.16 kDa and 7.73 kDa, respectively. Methylation with gas chromatography-mass spectrometry and NMR analyses confirmed that two fractions comprised of →4) -α-d- Glcp -(1→, →6) -β-d- Glcp -(1→ and →3,4) -β-d- Glcp -(1→) as the main chain. In addition, MPP-1 and MPP-2 polysaccharides showed significant antioxidant activity with respect to 1,1-diphenyl-2-picrylhydrazyl (DPPH), hydroxyl group and reducing power, and MPP-2 demonstrated excellent antioxidant activities compared to MPP-1 with IC50 values of 35.12, 18.82 and 40.12 μg mL-1, respectively. CONCLUSION The novel polysaccharide, MPP-1 and MPP-2, mainly containing glucose, showed significant antioxidant activity with respect to DPPH, hydroxyl group and reducing power. This study has enhanced waste utilization and reduced environmental pollution, providing some inspiration for the reuse of the waste generated during agricultural production. It may bring good prospects for their use as antioxidants in functional foods. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Gangjun Guo
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Wenting Xu
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Jiarong Fu
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Shangxuan Ma
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Kechang Huang
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Yuanmiao Wei
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Yuexue Yang
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Xiuhua Lan
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| | - Xiyong He
- Yunnan Institute of Tropical Crops, Jinghong, China
- Yunnan Macadamia Agricultural Engineering Research Center, Jinghong, China
| |
Collapse
|
4
|
Lv H, Miao M, Wu Z, Huang C, Tang X, Yan R. Hyaluronic acid-coated liposomes for enhanced in vivo efficacy of neogambogic acid via active tumor cell targeting and prolonged systemic exposure. J Liposome Res 2024; 34:605-616. [PMID: 38733152 DOI: 10.1080/08982104.2024.2348643] [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: 12/11/2023] [Revised: 04/15/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
Neogambogic acid (NGA), which possesses a variety of anticancer activities, is visualized as an anticancer bioactive ingredient. However, the huge vascular stimulation, poor aqueous solubility, and short half-life restricted its clinical use. In this work, an effective nanocarrier was explored to reduce toxicity and enhance the tumor-targeted delivery. Two liposomal formulations, neogambogic acid liposomes (NGA-L), and hyaluronic acid-coated neogambogic acid liposomes (HA-NGA-L) were prepared and characterized with high encapsulation efficiency, slow pattern of drug release, narrow size distribution and higher stability. The cytotoxicity and cellular uptake of HA-NGA-L were higher than those of NGA-L in MDA-MB-231 cells (high CD44 expression), while no obvious differences in MCF-7 cells with (low CD44 expression), suggesting the CD44-mediated cellular internalization of hyaluronic acid-modified liposomes enhanced the cytotoxicity. Mechanistically, elevation of Bax and caspase-3 as well as downregulation of Bcl-2 led to cell apoptosis. Besides, the vascular stimulation and the hemolysis test indicated good safety of HA-NGA-L. In addition, HA-NGA-L was the effective nanocarrier to repress tumor proliferation in MDA-MB-231 tumor xenograft mouse through CD44 mediated active targeting without any obvious histopathological abnormities on major organs. Immunohistochemistry analysis revealed the enhanced elevation of Bax and caspase-3, and reduced expression of Bcl-2 contribute to apoptosis in tumors. Meanwhile, HA-NGA-L increased the AUC and t1/2 by 5.34-fold and 3.94-fold, respectively. In summary, the present study shows that HA-NGA-L may be safe and effective for the tumor-targeted delivery of neogambogic acid.
Collapse
Affiliation(s)
- Hongzhen Lv
- School of Basic Medical Sciences, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Miao Miao
- Disease Prevention and Control Center of Tongshan District, Xuzhou, China
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhichao Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Cheng Huang
- School of Traditional Chinese Medicine, Jiangsu Vocational College of Medicine, Yancheng, China
| | - Xiaozhu Tang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rugen Yan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
5
|
Tanaka T, Sano K, Kawakami R, Tanaka S, Munekane M, Yamasaki T, Mukai T. Electrostatically self-assembled gold nanorods with sulfated hyaluronic acid for targeted photothermal therapy for CD44-positive tumors. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 62:102781. [PMID: 39163902 DOI: 10.1016/j.nano.2024.102781] [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: 04/09/2024] [Revised: 06/27/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024]
Abstract
Gold nanorods (GNR) produce heat upon irradiation with near-infrared light, enabling a tumor-targeted photothermal therapy. In this study, we prepared GNR coated with sulfated hyaluronic acid (sHA) with a binding affinity for CD44 via electrostatic interactions to deliver GNR to tumors efficiently and stably, and evaluated their usefulness for photothermal therapy. Cationic GNR modified with trimethylammonium groups electrostatically interacted with native HA or sHA with varying degrees of sulfation to form complexes. While GNR/HA was unstable in saline, GNR/sHA maintained the absorbance peak in the near-infrared region, particularly for GNR/sHA with higher degrees of sulfation. GNR/sHA exhibited an intense photothermal effect upon irradiation with near-infrared light. Furthermore, in vitro and in vivo studies revealed that GNR coated with sHA containing approximately 1.2 sulfated groups per HA unit could accumulate in CD44-positive tumors via an HA-specific pathway. These findings indicate the effectiveness of GNR/sHA as a tumor-targeted photothermal therapeutic agent.
Collapse
Affiliation(s)
- Toshie Tanaka
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Rin Kawakami
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Shiho Tanaka
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Masayuki Munekane
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Toshihide Yamasaki
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Takahiro Mukai
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan.
| |
Collapse
|
6
|
Zhao Y, Le TMD, Hong J, Jiao A, Yoon AR, Yun CO. Smart Accumulating Dual-Targeting Lipid Envelopes Equipping Oncolytic Adenovirus for Enhancing Cancer Gene Therapeutic Efficacy. ACS NANO 2024; 18:27869-27890. [PMID: 39356167 DOI: 10.1021/acsnano.4c02165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
Systemic delivery of oncolytic adenovirus (oAd) for cancer gene therapy must overcome several limitations such as rapid clearance from the blood, nonspecific accumulation in the liver, and insufficient delivery to the tumor tissues. In the present report, a tumor microenvironment-triggered artificial lipid envelope composed of a pH-responsive sulfamethazine-based polymer (PUSSM)-conjugated phospholipid (DOPE-HZ-PUSSM) and another lipid decorated with epidermal growth factor receptor (EGFR) targeting peptide (GE11) (GE11-DOPE) was utilized to encapsulate replication-incompetent Ad (dAd) or oAd coexpressing short-hairpin RNA (shRNA) against Wnt5 (shWnt5) and decorin (dAd/LP-GE-PS or oAd/LP-GE-PS, respectively). In vitro studies demonstrated that dAd/LP-GE-PS transduced breast cancer cells in a pH-responsive and EGFR-specific manner, showing a higher level of transduction than naked Ad under a mildly acidic pH of 6.0 in EGFR-positive cell lines. In vivo biodistribution analyses revealed that systemic administration of oAd/LP-GE-PS leads to a significantly higher level of intratumoral virion accumulation compared to naked oAd, oAd encapsulated in a liposome without PUSSM or EGFR targeting peptide moiety (oAd/LP), or oAd encapsulated in a liposome with EGFR targeting peptide alone (oAd/LP-GE) in an EGFR overexpressing MDA-MB-468 breast tumor xenograft model, showing that both pH sensitivity and EGFR targeting ability were integral to effective systemic delivery of oAd. Further, systemic administration of all liposomal oAd formulations (oAd/LP, oAd/LP-GE, and oAd/LP-GE-PS) showed significantly attenuated hepatic accumulation of the virus compared to naked oAd. Collectively, our findings demonstrated that pH-sensitive and EGFR-targeted liposomal systemic delivery of oAd can be a promising strategy to address the conventional limitations of oAd to effectively treat EGFR-positive cancer in a safe manner.
Collapse
Affiliation(s)
- Yuebin Zhao
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - Thai Minh Duy Le
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - Jinwoo Hong
- GeneMedicine CO., Ltd., Seoul 04763, South Korea
| | - Ao Jiao
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul 04763, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, South Korea
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul 04763, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul 04763, South Korea
- GeneMedicine CO., Ltd., Seoul 04763, South Korea
| |
Collapse
|
7
|
Wang Z, Pang S, Liu X, Dong Z, Tian Y, Ashrafizadeh M, Rabiee N, Ertas YN, Mao Y. Chitosan- and hyaluronic acid-based nanoarchitectures in phototherapy: Combination cancer chemotherapy, immunotherapy and gene therapy. Int J Biol Macromol 2024; 273:132579. [PMID: 38795895 DOI: 10.1016/j.ijbiomac.2024.132579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
Cancer phototherapy has been introduced as a new potential modality for tumor suppression. However, the efficacy of phototherapy has been limited due to a lack of targeted delivery of photosensitizers. Therefore, the application of biocompatible and multifunctional nanoparticles in phototherapy is appreciated. Chitosan (CS) as a cationic polymer and hyaluronic acid (HA) as a CD44-targeting agent are two widely utilized polymers in nanoparticle synthesis and functionalization. The current review focuses on the application of HA and CS nanostructures in cancer phototherapy. These nanocarriers can be used in phototherapy to induce hyperthermia and singlet oxygen generation for tumor ablation. CS and HA can be used for the synthesis of nanostructures, or they can functionalize other kinds of nanostructures used for phototherapy, such as gold nanorods. The HA and CS nanostructures can combine chemotherapy or immunotherapy with phototherapy to augment tumor suppression. Moreover, the CS nanostructures can be functionalized with HA for specific cancer phototherapy. The CS and HA nanostructures promote the cellular uptake of genes and photosensitizers to facilitate gene therapy and phototherapy. Such nanostructures specifically stimulate phototherapy at the tumor site, with particle toxic impacts on normal cells. Moreover, CS and HA nanostructures demonstrate high biocompatibility for further clinical applications.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Neurosurgery, Liaocheng Traditional Chinese Medicine Hospital, Liaocheng 252000, Shandong, PR China
| | - Shuo Pang
- Department of Urinary Surgery, Jinan Third People's Hospital, Jinan, Shandong 250101, PR China
| | - Xiaoli Liu
- Department of Dermatology, First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zi Dong
- Department of Gastroenterology, Lincang People's Hospital, Lincang, China
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, United States
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518055, China; International Association for Diagnosis and Treatment of Cancer, Shenzhen, Guangdong 518055, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China.
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, 600077 India
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri 38039, Türkiye; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri 38039, Türkiye; UNAM-National Nanotechnology Research Center, Bilkent University, Ankara 06800, Türkiye.
| | - Ying Mao
- Department of Oncology, Suining Central Hospital, Suining City, Sichuan, China.
| |
Collapse
|
8
|
Urbano-Gámez JD, Guzzi C, Bernal M, Solivera J, Martínez-Zubiaurre I, Caro C, García-Martín ML. Tumor versus Tumor Cell Targeting in Metal-Based Nanoparticles for Cancer Theranostics. Int J Mol Sci 2024; 25:5213. [PMID: 38791253 PMCID: PMC11121233 DOI: 10.3390/ijms25105213] [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: 04/11/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The application of metal-based nanoparticles (mNPs) in cancer therapy and diagnostics (theranostics) has been a hot research topic since the early days of nanotechnology, becoming even more relevant in recent years. However, the clinical translation of this technology has been notably poor, with one of the main reasons being a lack of understanding of the disease and conceptual errors in the design of mNPs. Strikingly, throughout the reported studies to date on in vivo experiments, the concepts of "tumor targeting" and "tumor cell targeting" are often intertwined, particularly in the context of active targeting. These misconceptions may lead to design flaws, resulting in failed theranostic strategies. In the context of mNPs, tumor targeting can be described as the process by which mNPs reach the tumor mass (as a tissue), while tumor cell targeting refers to the specific interaction of mNPs with tumor cells once they have reached the tumor tissue. In this review, we conduct a critical analysis of key challenges that must be addressed for the successful targeting of either tumor tissue or cancer cells within the tumor tissue. Additionally, we explore essential features necessary for the smart design of theranostic mNPs, where 'smart design' refers to the process involving advanced consideration of the physicochemical features of the mNPs, targeting motifs, and physiological barriers that must be overcome for successful tumor targeting and/or tumor cell targeting.
Collapse
Affiliation(s)
- Jesús David Urbano-Gámez
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Cinzia Guzzi
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - Manuel Bernal
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, 29071 Malaga, Spain
| | - Juan Solivera
- Department of Neurosurgery, Reina Sofia University Hospital, 14004 Cordoba, Spain;
| | - Iñigo Martínez-Zubiaurre
- Department of Clinical Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, P.O. Box 6050, Langnes, 9037 Tromsö, Norway;
| | - Carlos Caro
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
| | - María Luisa García-Martín
- Biomedical Magnetic Resonance Laboratory—BMRL, Andalusian Public Foundation Progress and Health—FPS, 41092 Seville, Spain; (J.D.U.-G.); (C.G.)
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma BIONAND, C/Severo Ochoa, 35, 29590 Malaga, Spain;
- Biomedical Research Networking Center in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
9
|
Liu H, Zhang M, Meng F, Wubuli A, Li S, Xiao S, Gu L, Li J. HAuCl 4-mediated green synthesis of highly stable Au NPs from natural active polysaccharides: Synthetic mechanism and antioxidant property. Int J Biol Macromol 2024; 265:130824. [PMID: 38492708 DOI: 10.1016/j.ijbiomac.2024.130824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Polysaccharide-functionalized gold nanoparticles (Polysaccharide-Au NPs) with high stability were successfully prepared by a straightforward method. Notably, the Au (III) ion acts as a strong Lewis acid to facilitate glycosidic bond breaking. Subsequently, the polysaccharide conformation was transformed to an open-chain form, exposing highly reduced aldehyde or ketone groups that reduce Au (III) to Au (0) crystal species, further growing into Au NPs. As-prepared Au NPs displayed excellent stability over a longer storage period (more than 70 days), a wide range of temperatures (25-60 °C), and pH range (3-11), varying concentrations (0-200 mM) and types of salt ions (Na+, K+, Ca2+, Mg2+), and glutathione solutions (5 mM). More interestingly, polysaccharide-Au NPs retained the antioxidant activity of polysaccharides and reduced oxidative damage at the cellular level through decreased reactive oxygen species (ROS) production. The intracellular levels of ROS pretreated with polysaccharide and polysaccharide-Au NPs were decreased 53.12-75.85 % compared to the H2O2 group, respectively. Therefore, the green synthesized Au NPs from natural active polysaccharides exhibit potential applications in biomedical fields.
Collapse
Affiliation(s)
- Haoqiang Liu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Minwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Fanxing Meng
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Abudukahaer Wubuli
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Suxin Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Shuang Xiao
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Liyu Gu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
10
|
Tang Z, Hou Y, Huang S, Hosmane NS, Cui M, Li X, Suhail M, Zhang H, Ge J, Iqbal MZ, Kong X. Dumbbell-shaped bimetallic AuPd nanoenzymes for NIR-II cascade catalysis-photothermal synergistic therapy. Acta Biomater 2024; 177:431-443. [PMID: 38307478 DOI: 10.1016/j.actbio.2024.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024]
Abstract
The noble metal NPs that are currently applied to photothermal therapy (PTT) have their photoexcitation location mainly in the NIR-I range, and the low tissue penetration limits their therapeutic effect. The complexity of the tumor microenvironment (TME) makes it difficult to inhibit tumor growth completely with a single therapy. Although TME has a high level of H2O2, the intratumor H2O2 content is still insufficient to catalyze the generation of sufficient hydroxide radicals (‧OH) to achieve satisfactory therapeutic effects. The AuPd-GOx-HA (APGH) was obtained from AuPd bimetallic nanodumbbells modified by glucose oxidase (GOx) and hyaluronic acid (HA) for photothermal enhancement of tumor starvation and cascade catalytic therapy in the NIR-II region. The CAT-like activity of AuPd alleviates tumor hypoxia by catalyzing the decomposition of H2O2 into O2. The GOx-mediated intratumoral glucose oxidation on the one hand can block the supply of energy and nutrients essential for tumor growth, leading to tumor starvation. On the other hand, the generated H2O2 can continuously supply local O2, which also exacerbates glucose depletion. The peroxidase-like activity of bimetallic AuPd can catalyze the production of toxic ‧OH radicals from H2O2, enabling cascade catalytic therapy. In addition, the high photothermal conversion efficiency (η = 50.7 %) of APGH nanosystems offers the possibility of photothermal imaging-guided photothermal therapy. The results of cell and animal experiments verified that APGH has good biosafety, tumor targeting, and anticancer effects, and is a precious metal nanotherapeutic system integrating glucose starvation therapy, nano enzyme cascade catalytic therapy, and PTT therapy. This study provides a strategy for photothermal-cascade catalytic synergistic therapy combining both exogenous and endogenous processes. STATEMENT OF SIGNIFICANCE: AuPd-GOx-HA cascade nanoenzymes were prepared as a potent cascade catalytic therapeutic agent, which enhanced glucose depletion, exacerbated tumor starvation and promoted cancer cell apoptosis by increasing ROS production through APGH-like POD activity. The designed system has promising photothermal conversion ability in the NIR-II region, simultaneously realizing photothermal-enhanced catalysis, PTT, and catalysis/PTT synergistic therapy both in vitro and in vivo. The present work provides an approach for designing and developing catalytic-photothermal therapies based on bimetallic nanoenzymatic cascades.
Collapse
Affiliation(s)
- Zhe Tang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yike Hou
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Shuqi Huang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Narayan S Hosmane
- Department of Chemistry & Biochemistry, Northern Illinois University, DeKalb, IL 60115, USA
| | - Mingyue Cui
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xianan Li
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Muhammad Suhail
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Han Zhang
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Jian Ge
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone, Hangzhou 310018, China
| | - M Zubair Iqbal
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| | - Xiangdong Kong
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
11
|
Wang B, Xu XJ, Fu Y, Ren B, Yang XD, Yang HY. A tumor-targeted and enzyme-responsive gold nanorod-based nanoplatform with facilitated endo-lysosomal escape for synergetic photothermal therapy and protein therapy. Dalton Trans 2024; 53:2120-2130. [PMID: 38180436 DOI: 10.1039/d3dt03305h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
To tackle the obstacles related to tumor targeting and overcome the limitations of single treatment models, we have developed a nanoplatform that is both tumor-targeted and enzyme-responsive. This nanoplatform integrates photothermal gold nanorods (AuNRs) and protein drugs into a single system. This nanosystem, known as AuNRs@HA-mPEG-Deta-LA, was fabricated by modifying gold nanorods (AuNRs) with a polymeric ligand called hyaluronic acid-grafted-(mPEG/diethylenetriamine-conjugated-lipoic acid). The purpose of this fabrication was to load cytochrome c (CC) and utilize it for the synergetic protein-photothermal therapy of cancer. The resulting nanoplatform exhibited a high efficiency in loading proteins and demonstrated excellent stability in different biological environments. Additionally, CC-loaded AuNRs@HA-mPEG-Deta-LA not only enabled localized hyperthermia for photothermal therapy (PTT) with laser irradiation but also facilitated the release of CC under the action of hyaluronidase, an enzyme known to be overexpressed in tumor cells. The confocal imaging results demonstrated that the presence of a specific polymeric ligand on this nanoparticle enhances the internalization of CD44-positive cancer cells, accelerates endo/lysosomal escape, and facilitates the controlled release of CC within the cells. Furthermore, the results of the MTT assay also showed that AuNRs@HA-mPEG-Deta-LA as a protein nanocarrier demonstrated excellent biocompatibility. Importantly, this synergistic therapeutic strategy effectively induced apoptosis in A549 cancer cells by increasing the intracellular concentration of CC and utilizing the photothermal conversion of AuNRs, which was observed to be more effective compared to using only protein therapy or PTT. Therefore, this study showcased a nanoplatform based on AuNRs that has great potential for tumor-targeted protein delivery in combination with PTT in cancer treatment.
Collapse
Affiliation(s)
- Bo Wang
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Xin Jun Xu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| | - Yan Fu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| | - Bo Ren
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Xiao Dong Yang
- Jilin Provincial Key Laboratory of Straw-Based Functional Materials, Institute for Interdisciplinary Biomass Functional Materials Studies, Jilin Engineering Normal University, Changchun 130052, P. R. China.
| | - Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| |
Collapse
|
12
|
Liu H, Zhang M, Meng F, Su C, Li J. Polysaccharide-based gold nanomaterials: Synthesis mechanism, polysaccharide structure-effect, and anticancer activity. Carbohydr Polym 2023; 321:121284. [PMID: 37739497 DOI: 10.1016/j.carbpol.2023.121284] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 09/24/2023]
Abstract
Polysaccharide-based gold nanomaterials have attracted great interest in biomedical fields such as cancer therapy and immunomodulation due to their prolonged residence time in vivo and enhanced immune response. This review aims to provide an up-to-date and comprehensive summary of polysaccharide-based Au NMs synthesis, including mechanisms, polysaccharide structure-effects, and anticancer activity. Firstly, research progress on the synthesis mechanism of polysaccharide-based Au NMs was addressed, which included three types based on the variety of polysaccharides and reaction environment: breaking of glycosidic bonds via Au (III) or base-mediated production of highly reduced intermediates, reduction of free hydroxyl groups in polysaccharide molecules, and reduction of free amino groups in polysaccharide molecules. Then, the potential effects of polysaccharide structure characteristics (molecular weight, composition of monosaccharides, functional groups, glycosidic bonds, and chain conformation) and reaction conditions (the reaction temperature, reaction time, pH, concentration of gold precursor and polysaccharides) on the size and shape of Au NMs were explored. Finally, the current status of polysaccharide-based Au NMs cancer therapy was summarized before reaching our conclusions and perspectives.
Collapse
Affiliation(s)
- Haoqiang Liu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Minwei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Fanxing Meng
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Chenyi Su
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science & Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
13
|
Yang M, Zhang Y, Hu Z, Xie H, Tian W, Liu Z. Application of hyaluronic acid-based nanoparticles for cancer combination therapy. Int J Pharm 2023; 646:123459. [PMID: 37778513 DOI: 10.1016/j.ijpharm.2023.123459] [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: 05/07/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Cancer is a significant public health problem in the world. The treatment methods include surgery, chemotherapy, phototherapy, and immunotherapy. Due to their respective limitations, the treatment effect is often unsatisfactory, laying hidden dangers for metastasis and recurrence. Since their exceptional biocompatibility and excellent targeting capabilities, hyaluronic acid-based biomaterials have generated great interest as drug delivery methods for tumor therapy. Moreover, modified HA can self-assemble into hydrogels or nanoparticles (NPs) for precise drug administration. This article summarizes the application of HA-based NPs in combination therapy. Ultimately, it is anticipated that this research will offer guidance for creating various HA-based NPs utilized in numerous cancer therapies.
Collapse
Affiliation(s)
- Mengru Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Ying Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zheming Hu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Haonan Xie
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Wenli Tian
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China
| | - Zhidong Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
14
|
Ju J, Xu D, Mo X, Miao J, Xu L, Ge G, Zhu X, Deng H. Multifunctional polysaccharide nanoprobes for biological imaging. Carbohydr Polym 2023; 317:121048. [PMID: 37364948 DOI: 10.1016/j.carbpol.2023.121048] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/28/2023]
Abstract
Imaging and tracking biological targets or processes play an important role in revealing molecular mechanisms and disease states. Bioimaging via optical, nuclear, or magnetic resonance techniques enables high resolution, high sensitivity, and high depth imaging from the whole animal down to single cells via advanced functional nanoprobes. To overcome the limitations of single-modality imaging, multimodality nanoprobes have been engineered with a variety of imaging modalities and functionalities. Polysaccharides are sugar-containing bioactive polymers with superior biocompatibility, biodegradability, and solubility. The combination of polysaccharides with single or multiple contrast agents facilitates the development of novel nanoprobes with enhanced functions for biological imaging. Nanoprobes constructed with clinically applicable polysaccharides and contrast agents hold great potential for clinical translations. This review briefly introduces the basics of different imaging modalities and polysaccharides, then summarizes the recent progress of polysaccharide-based nanoprobes for biological imaging in various diseases, emphasizing bioimaging with optical, nuclear, and magnetic resonance techniques. The current issues and future directions regarding the development and applications of polysaccharide nanoprobes are further discussed.
Collapse
Affiliation(s)
- Jingxuan Ju
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Danni Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuan Mo
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiaqian Miao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Xu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Hongping Deng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
15
|
Wang L, Shrestha B, Brey EM, Tang L. Gold Nanomaterial System That Enables Dual Photothermal and Chemotherapy for Breast Cancer. Pharmaceutics 2023; 15:2198. [PMID: 37765168 PMCID: PMC10534904 DOI: 10.3390/pharmaceutics15092198] [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: 08/01/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
This study involves the fabrication and characterization of a multifunctional therapeutic nanocomposite system, as well as an assessment of its in vitro efficacy for breast cancer treatment. The nanocomposite system combines gold nanorods (GNRs) and gold nanoclusters (GNCs) to enable a combination of photothermal therapy and doxorubicin-based chemotherapy. GNRs of various sizes but exhibiting similar absorbance spectra were synthesized and screened for photothermal efficiency. GNRs exhibiting the highest photothermal efficiency were selected for further experiments. GNCs were synthesized in bovine serum albumin (BSA) and integrated into citrate-capped GNRs using layer-by-layer assembly. Glutaraldehyde crosslinking with the lysine residues in BSA was employed to immobilize the GNCs onto the GNRs, forming a stable "soft gel-like" structure. This structure provided binding sites for doxorubicin through electrostatic interactions and enhanced the overall structural stability of the nanocomposite. Additionally, the presence of GNCs allowed the nanocomposite system to emit robust fluorescence in the range of ~520 nm to 700 nm for self-detection. Hyaluronic acid was functionalized on the exterior surface of the nanocomposite as a targeting moiety for CD44 to improve the cellular internalization and specificity for breast cancer cells. The developed nanocomposite system demonstrated good stability in vitro and exhibited a pH- and near-infrared-responsive drug release behavior. In vitro studies showed the efficient internalization of the nanocomposite system and reduced cellular viability following NIR irradiation in MDA-MB-231 breast cancer cells. Together, these results highlight the potential of this nanocomposite system for targeted breast cancer therapy.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Biomedical Engineering & Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, USA;
| | - Binita Shrestha
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78705, USA
| | - Eric M. Brey
- Department of Biomedical Engineering & Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, USA;
| | - Liang Tang
- Department of Biomedical Engineering & Chemical Engineering, University of Texas at San Antonio, San Antonio, TX 78249, USA;
| |
Collapse
|
16
|
Jiang Z, Jiang Z, Jiang Y, Cheng Y, Yao Q, Chen R, Kou L. Fe-involved nanostructures act as photothermal transduction agents in cancer photothermal therapy. Colloids Surf B Biointerfaces 2023; 228:113438. [PMID: 37421763 DOI: 10.1016/j.colsurfb.2023.113438] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
Cancer, a disease notorious for its difficult therapy regimen, has long puzzled researchers. Despite attempts to cure cancer using surgery, chemotherapy, radiotherapy, and immunotherapy, their effectiveness is limited. Recently, photothermal therapy (PTT), a rising strategy, has gained attention. PTT can increase the surrounding temperature of cancer tissues and cause damage to them. Fe is widely used in PTT nanostructures due to its strong chelating ability, good biocompatibility, and the potential to induce ferroptosis. In recent years, many nanostructures incorporating Fe3+ have been developed. In this article, we summarize PTT nanostructures containing Fe and introduce their synthesis and therapy strategy. However, PTT nanostructures containing Fe are still in their infancy, and more effort must be devoted to improving their effectiveness so that they can eventually be used in clinics.
Collapse
Affiliation(s)
- Zewei Jiang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Zhikai Jiang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yiling Jiang
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yingfeng Cheng
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China
| | - Qing Yao
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Ruijie Chen
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China.
| | - Longfa Kou
- Wenzhou Municipal Key Laboratory of Pediatric Pharmacy, Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou 325027, China; Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Wenzhou 325000, China; Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou 325000, China.
| |
Collapse
|
17
|
Uthappa UT, Suneetha M, Ajeya KV, Ji SM. Hyaluronic Acid Modified Metal Nanoparticles and Their Derived Substituents for Cancer Therapy: A Review. Pharmaceutics 2023; 15:1713. [PMID: 37376161 DOI: 10.3390/pharmaceutics15061713] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/17/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
The use of metal nanoparticles (M-NPs) in cancer therapy has gained significant consideration owing to their exceptional physical and chemical features. However, due to the limitations, such as specificity and toxicity towards healthy cells, their application in clinical translations has been restricted. Hyaluronic acid (HA), a biocompatible and biodegradable polysaccharide, has been extensively used as a targeting moiety, due to its ability to selectively bind to the CD44 receptors overexpressed on cancer cells. The HA-modified M-NPs have demonstrated promising results in improving specificity and efficacy in cancer therapy. This review discusses the significance of nanotechnology, the state of cancers, and the functions of HA-modified M-NPs, and other substituents in cancer therapy applications. Additionally, the role of various types of selected noble and non-noble M-NPs used in cancer therapy are described, along with the mechanisms involved in cancer targeting. Additionally, the purpose of HA, its sources and production processes, as well as its chemical and biological properties are described. In-depth explanations are provided about the contemporary applications of HA-modified noble and non-noble M-NPs and other substituents in cancer therapy. Furthermore, potential obstacles in optimizing HA-modified M-NPs, in terms of clinical translations, are discussed, followed by a conclusion and future prospects.
Collapse
Affiliation(s)
- Uluvangada Thammaiah Uthappa
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
- Department of Bioengineering, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, India
| | - Maduru Suneetha
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Kanalli V Ajeya
- Department of Environment and Energy Engineering, Chonnam National University, 77 Yongbong-Ro, Buk-gu, Gwangju 61186, Republic of Korea
| | - Seong Min Ji
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
18
|
Yang B, Yin S, Zhou Z, Huang L, Xi M. Inflammation Control and Tumor Growth Inhibition of Ovarian Cancer by Targeting Adhesion Molecules of E-Selectin. Cancers (Basel) 2023; 15:cancers15072136. [PMID: 37046797 PMCID: PMC10093113 DOI: 10.3390/cancers15072136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/27/2023] [Accepted: 04/02/2023] [Indexed: 04/07/2023] Open
Abstract
Objective: The aim is to use E-selectin-binding peptide (ESBP) to actively recognize E-selectin, so allowing a drug delivery system to actively recognize the cells and inhibit the tumor growth of ovarian cancer by targeting adhesion molecules of E-selectin. An ovarian-cancer-directed drug delivery system was designed based on the high affinity of E-selectin-binding peptide (ESBP) to E-selectin. The effects and mechanisms of ESBP-bovine serum albumin (BSA) polymerized nanoparticles were investigated. Methods: BSA polymerized nanoparticles (BSANPs) and ESBP-BSANPs-paclitaxel (PTX) were prepared and their characteristics were measured. The in vitro targetability and cytotoxicity of ESBP-BSANPs-PTX were evaluated through in vitro drug uptake and MTT experiments. The mechanisms of ESBP-BSANPs-PTX were investigated via apoptosis, wound healing and immunohistochemistry assays. The in vivo targeting properties and drug effects were observed in a mouse tumor-bearing model. Results: In vitro experiments revealed an increase in the uptake of ESBP-BSANPs-FITC. The cytotoxicity of ESBP-BSANPs-PTX in A2780/CP70, HUVEC, RAW264.7 and ID8 cells was higher than that of PTX alone. ESBP-BSANPs-PTX increased cell apoptosis in a dose-dependent manner and exhibited a greater ability to inhibit cell migration than BSANPs-PTX. In vivo experiments demonstrated the targetability and good effects of ESBP-BSANPs. Conclusions: ESBP-BSANPs-PTX improve PTX targetability, provide tumor-specific and potent therapeutic activities, and show promise for the development of agents in preclinical epithelial ovarian cancer.
Collapse
Affiliation(s)
- Bowen Yang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| | - Shanmei Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zishuo Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Luyao Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Mingrong Xi
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
| |
Collapse
|
19
|
Wang J, Wu Y, Liu K, Yang W, Zeng W, Gao X, Liu S, Zhang B. pH/thermal dual-responsive multifunctional drug delivery system for effective photoacoustic imaging-guided tumor chemo/photothermal therapy. APL Bioeng 2023; 7:016115. [PMID: 36974040 PMCID: PMC10039759 DOI: 10.1063/5.0139929] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/02/2023] [Indexed: 03/29/2023] Open
Abstract
The development of a combination of chemo/photothermal therapy could overcome the limitations of single-modality therapy and enhance therapeutic efficacy. In this study, a pH/thermal dual-responsive multifunctional drug delivery system with dual-drug loading and enhanced chemo/photothermal therapy is developed based on polydopamine-coated mesoporous silica-gold nanorods (PDA-AuNRs@MSN). Nanoscale mesoporous silica-gold nanorods encapsulating doxorubicin (DOX) are designed as a core and then modified by polydopamine. The PDA shell not only conjugates with another anticancer bortezomib (Btz) to form pH-sensitive bond through boronic acid and catechol but also acts as a gatekeeper to control the release of doxorubicin and enhance the photothermal effect. Such a nanocarrier not only acts as a contrast agent for PA imaging but also serves as a therapeutic agent for enhanced chemo/photothermal therapy. The DOX and Btz could be released in an on-demand mode under near-infrared light irradiation and acid environment. The tumor size and location could be observed via PA imaging after intravenous injection into 4T1-bearing mice. Compared with AuNRs@MSN, PDA-AuNRs@MSN exhibit an increased near-infrared (NIR) absorption at 808 nm and an enhanced photothermal effect. The integrated D/B-PDA-AuNRs@MSN nanoparticles show higher cell apoptosis and enhanced tumor treatment efficacy in vitro and in vivo in comparison with single chemotherapy or photothermal therapy. Combined together, D/B-PDA-AuNRs@MSN show pH/thermal-responsive controlled-release and synergistic chemo/photothermal therapy for tumor.
Collapse
Affiliation(s)
- Jun Wang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - YanYan Wu
- Department of Radiology, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Kai Liu
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Weitao Yang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Weiwei Zeng
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xiaolong Gao
- Department of Radiology, Luodian Hospital, Baoshan District, Shanghai 201908, China
| | - ShiYuan Liu
- Department of Radiology, The Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
- Authors to whom correspondence should be addressed: and
| | - Bingbo Zhang
- Department of Radiology, Tongji Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, The Institute for Biomedical Engineering and Nano Science, School of Medicine, Tongji University, Shanghai 200065, China
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
20
|
Ding YF, Xu X, Li J, Wang Z, Luo J, Mok GSP, Li S, Wang R. Hyaluronic Acid-based Supramolecular Nanomedicine with Optimized Ratio of Oxaliplatin/Chlorin e6 for Combined Chemotherapy and O2-Economized Photodynamic Therapy. Acta Biomater 2023; 164:397-406. [PMID: 37004784 DOI: 10.1016/j.actbio.2023.03.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/23/2023] [Accepted: 03/26/2023] [Indexed: 04/03/2023]
Abstract
Dual- or multi-modality combination therapy has become one of the most effective strategies to overcome drug resistance in cancer therapy, and the optimized ratio of the therapeutic agents working on the tumor greatly affects the therapeutic outcomes. However, the absence of a facile method to optimize the ratio of therapeutic agents in nanomedicine has, at least in part, impaired the clinical potential of combination therapy. Herein, a new cucurbit[7]uril (CB[7])-conjugated hyaluronic acid (HA) based nanomedicine was developed, in which both chlorin e6 (Ce6) and oxaliplatin (OX) were co-loaded non-covalently at an optimized ratio via facile host-guest complexation, for optimal, combined photodynamic therapy (PDT)/chemotherapy. To maximize the therapeutic efficacy, a mitochondrial respiration inhibitor, atovaquone (Ato), was also loaded into the nanomedicine to limit consumption of oxygen by the solid tumor, sparing oxygen for more efficient PDT. Additionally, HA on the surface of nanomedicine allowed targeted delivery to cancer cells with over-expressed CD44 receptors (such as CT26 cell lines). Thus, this supramolecular nanomedicine platform with an optimal ratio of photosensitizer and chemotherapeutic agent not only provides an important new tool for enhanced PDT/chemotherapy of solid tumors, but also offers a CB[7]-based host-guest complexation strategy to facilely optimize the ratio of therapeutic agents for multi-modality nanomedicine. STATEMENT OF SIGNIFICANCE: Chemotherapy remains the most common modality for cancer treatment in clinical practice. Combination therapy by co-delivery of two or more therapeutic agents has been recognized as one of the most effective strategies to improve therapeutic outcome of cancer treatment. However, the ratio of loaded drugs could not be facilely optimized, which may greatly affect the combination efficiency and overall therapeutic outcome. Herein, we developed a hyaluronic acid based supramolecular nanomedicine with facile method to optimize the ratio of two therapeutic agents for improved therapeutic outcome. This supramolecular nanomedicine not only provides an important new tool for enhanced photodynamic therapy/chemotherapy of solid tumors, but also offers insights in using macrocyclic molecule-based host-guest complexation to facilely optimize the ratio of therapeutic agents in multi-modality nanomedicine.
Collapse
Affiliation(s)
- Yuan-Fu Ding
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China; Biomedical Imaging Laboratory (BIG), Department of Electrical and Computer Engineering, University of Macau, Taipa, Macau SAR, China
| | - Xun Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Junyan Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | - Ziyi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China
| | | | - Greta S P Mok
- Biomedical Imaging Laboratory (BIG), Department of Electrical and Computer Engineering, University of Macau, Taipa, Macau SAR, China
| | - Shengke Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China.
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau SAR, China; MoE Frontier Centre for Precision Oncology, University of Macau, Taipa, Macau SAR, China.
| |
Collapse
|
21
|
Soni A, Bhandari MP, Tripathi GK, Bundela P, Khiriya PK, Khare PS, Kashyap MK, Dey A, Vellingiri B, Sundaramurthy S, Suresh A, Pérez de la Lastra JM. Nano-biotechnology in tumour and cancerous disease: A perspective review. J Cell Mol Med 2023; 27:737-762. [PMID: 36840363 PMCID: PMC10002932 DOI: 10.1111/jcmm.17677] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/07/2022] [Accepted: 11/18/2022] [Indexed: 02/26/2023] Open
Abstract
In recent years, drug manufacturers and researchers have begun to consider the nanobiotechnology approach to improve the drug delivery system for tumour and cancer diseases. In this article, we review current strategies to improve tumour and cancer drug delivery, which mainly focuses on sustaining biocompatibility, biodistribution, and active targeting. The conventional therapy using cornerstone drugs such as fludarabine, cisplatin etoposide, and paclitaxel has its own challenges especially not being able to discriminate between tumour versus normal cells which eventually led to toxicity and side effects in the patients. In contrast to the conventional approach, nanoparticle-based drug delivery provides target-specific delivery and controlled release of the drug, which provides a better therapeutic window for treatment options by focusing on the eradication of diseased cells via active targeting and sparing normal cells via passive targeting. Additionally, treatment of tumours associated with the brain is hampered by the impermeability of the blood-brain barriers to the drugs, which eventually led to poor survival in the patients. Nanoparticle-based therapy offers superior delivery of drugs to the target by breaching the blood-brain barriers. Herein, we provide an overview of the properties of nanoparticles that are crucial for nanotechnology applications. We address the potential future applications of nanobiotechnology targeting specific or desired areas. In particular, the use of nanomaterials, biostructures, and drug delivery methods for the targeted treatment of tumours and cancer are explored.
Collapse
Affiliation(s)
- Ambikesh Soni
- School of NanotechnologyRajiv Gandhi Proudyogiki VishwavidyalayaBhopalIndia
| | | | | | - Priyavand Bundela
- School of NanotechnologyRajiv Gandhi Proudyogiki VishwavidyalayaBhopalIndia
| | | | | | - Manoj Kumar Kashyap
- Amity Stem Cell Institute, Amity Medical SchoolAmity University HaryanaHaryanaIndia
| | - Abhijit Dey
- Department of Life SciencesPresidency UniversityWest BengalKolkataIndia
| | - Balachandar Vellingiri
- Stem cell and Regenerative Medicine/Translational ResearchDepartment of ZoologySchool of Basic Sciences, Central University of PunjabMaulana Azad National Institute of TechnologyBathindaIndia
| | - Suresh Sundaramurthy
- Department of Chemical EngineeringMaulana Azad National Institute of TechnologyMadhya PradeshBhopalIndia
| | - Arisutha Suresh
- Department of EnergyMaulana Azad National Institute of Technology & M/s Eco Science & TechnologyMadhya PradeshBhopalIndia
| | - José M. Pérez de la Lastra
- Biotecnología de macromoléculasInstituto de Productos Naturales y Agrobiología, (IPNA‐CSIC)San Cristóbal de la LagunaSpain
| |
Collapse
|
22
|
Li Z, Wang Q, Huang X, Yang M, Zhou S, Li Z, Fang Z, Tang Y, Chen Q, Hou H, Li L, Fei F, Wang Q, Wu Y, Gong A. Lactate in the tumor microenvironment: A rising star for targeted tumor therapy. Front Nutr 2023; 10:1113739. [PMID: 36875841 PMCID: PMC9978120 DOI: 10.3389/fnut.2023.1113739] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Metabolic reprogramming is one of fourteen hallmarks of tumor cells, among which aerobic glycolysis, often known as the "Warburg effect," is essential to the fast proliferation and aggressive metastasis of tumor cells. Lactate, on the other hand, as a ubiquitous molecule in the tumor microenvironment (TME), is generated primarily by tumor cells undergoing glycolysis. To prevent intracellular acidification, malignant cells often remove lactate along with H+, yet the acidification of TME is inevitable. Not only does the highly concentrated lactate within the TME serve as a substrate to supply energy to the malignant cells, but it also works as a signal to activate multiple pathways that enhance tumor metastasis and invasion, intratumoral angiogenesis, as well as immune escape. In this review, we aim to discuss the latest findings on lactate metabolism in tumor cells, particularly the capacity of extracellular lactate to influence cells in the tumor microenvironment. In addition, we examine current treatment techniques employing existing medications that target and interfere with lactate generation and transport in cancer therapy. New research shows that targeting lactate metabolism, lactate-regulated cells, and lactate action pathways are viable cancer therapy strategies.
Collapse
Affiliation(s)
- Zhangzuo Li
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Xufeng Huang
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Mengting Yang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shujing Zhou
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zhengrui Li
- School of Medicine, College of Stomatology, Shanghai Jiao Tong University, Shanghai, China.,National Center for Stomatology and National Clinical Research Center for Oral Diseases, Shanghai, China.,Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Zhengzou Fang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yidan Tang
- Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Qian Chen
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hanjin Hou
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Li
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Fei Fei
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiaowei Wang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuqing Wu
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Aihua Gong
- Hematological Disease Institute of Jiangsu University, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China.,Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
23
|
Wang XY, Lin C, Chang WJ, Huang YH, Mi FL. Thiolated hyaluronic acid and catalase-enhanced CD44-targeting and oxygen self-supplying nanoplatforms with photothermal/photodynamic effects against hypoxic breast cancer cells. Int J Biol Macromol 2022; 221:121-134. [PMID: 36049568 DOI: 10.1016/j.ijbiomac.2022.08.164] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/02/2022] [Accepted: 08/24/2022] [Indexed: 12/16/2022]
Abstract
Photothermal and photodynamic therapies (PTT/PDT) have been widely accepted as noninvasive therapeutic methods for cancer treatment. However, tumor hypoxia and insufficient delivery of photoactive compounds to cancer cells can reduce the efficacy of phototherapy. Herein, we first synthesized thiolated hyaluronic acid (THA) and then conjugated it with catalase (CAT) onto chlorin e6 (Ce6)-adsorbed small gold nanorods (Ce6@sAuNRs) with near-infrared (NIR)/visible light activated photothermal/photodynamic effects. The conjugation of THA and CAT on Ce6@sAuNRs resulted in a red-shift of the longitudinal LSPR absorption band of sAuNRs up to 1000 nm and maintained the excellent enzymatic activity of catalase. Modification of Ce6@sAuNRs with THA resulted in efficient internalization of the nanocomposite into MCF-7/ADR multidrug-resistant (MDR) breast cancer cells (CD44+), thereby significantly enhancing the intracellular accumulation of the photosensitizer Ce6. CAT endows Ce6@sAuNRs with self-supporting oxygen production, which enables them to efficiently generate singlet oxygen (1O2) under 660 nm laser irradiation and enhances the photodynamic effect against hypoxic breast cancer cells. The results highlight the prospect of this novel multi-functional nanoplatform integrating active biological macromolecules (THA and CAT) into photosensitizer/photothermal gold nanocomposites in overcoming the limitations of hypoxic MDR breast cancer cell treatment.
Collapse
Affiliation(s)
- Xin-Yu Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
| | - Chi Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
| | - Wong-Jin Chang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
| | - Yen-Hua Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan; International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Fwu-Long Mi
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan; International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City 11031, Taiwan.
| |
Collapse
|
24
|
Gold Nanorod-Assisted Photothermal Therapy and Improvement Strategies. Bioengineering (Basel) 2022; 9:bioengineering9050200. [PMID: 35621478 PMCID: PMC9138169 DOI: 10.3390/bioengineering9050200] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 11/17/2022] Open
Abstract
Noble metal nanoparticles have been sought after in cancer nanomedicine during the past two decades, owing to the unique localized surface plasmon resonance that induces strong absorption and scattering properties of the nanoparticles. A popular application of noble metal nanoparticles is photothermal therapy, which destroys cancer cells by heat generated by laser irradiation of the nanoparticles. Gold nanorods have stood out as one of the major types of noble metal nanoparticles for photothermal therapy due to the facile tuning of their optical properties in the tissue penetrative near infrared region, strong photothermal conversion efficiency, and long blood circulation half-life after surface modification with stealthy polymers. In this review, we will summarize the optical properties of gold nanorods and their applications in photothermal therapy. We will also discuss the recent strategies to improve gold nanorod-assisted photothermal therapy through combination with chemotherapy and photodynamic therapy.
Collapse
|
25
|
Kuang Y, Zhai J, Xiao Q, Zhao S, Li C. Polysaccharide/mesoporous silica nanoparticle-based drug delivery systems: A review. Int J Biol Macromol 2021; 193:457-473. [PMID: 34710474 DOI: 10.1016/j.ijbiomac.2021.10.142] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/30/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022]
Abstract
Mesoporous silica nanoparticles (MSNs) have been well-researched in the design and fabrication of advanced drug delivery systems (DDSs) due to their advantages such as good biocompatibility, large specific surface area and pore volume for drug loading, easily surface modification, adjusted size and good thermal/chemical stability. For MSN-based DDSs, gate materials are also necessary. And natural polysaccharides, one kind of the most abundant natural resource, have been widely applied as the "gatekeepers" in MSN-based DDSs. Polysaccharides are cheap and rich in sources with good biocompatibility, and some of them have important biological functions. In this review article, polysaccharides including chitosan, hyaluronic acid, sodium alginate and dextran, et al. are briefly introduced. And the preparation processes and properties such as controlled drug release, cancer targeting and disease diagnosis of functional polysaccharide/MSN-based DDSs are discussed.
Collapse
Affiliation(s)
- Ying Kuang
- Glyn O. Philips Hydrocolloid Research Centre at HUT, Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Junjun Zhai
- Glyn O. Philips Hydrocolloid Research Centre at HUT, Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Qinjian Xiao
- Glyn O. Philips Hydrocolloid Research Centre at HUT, Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Si Zhao
- Glyn O. Philips Hydrocolloid Research Centre at HUT, Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Cao Li
- Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, Hubei Key Laboratory of Polymer Materials, Hubei University, Wuhan 430062, China.
| |
Collapse
|
26
|
Ashrafizadeh M, Mirzaei S, Gholami MH, Hashemi F, Zabolian A, Raei M, Hushmandi K, Zarrabi A, Voelcker NH, Aref AR, Hamblin MR, Varma RS, Samarghandian S, Arostegi IJ, Alzola M, Kumar AP, Thakur VK, Nabavi N, Makvandi P, Tay FR, Orive G. Hyaluronic acid-based nanoplatforms for Doxorubicin: A review of stimuli-responsive carriers, co-delivery and resistance suppression. Carbohydr Polym 2021; 272:118491. [PMID: 34420747 DOI: 10.1016/j.carbpol.2021.118491] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022]
Abstract
An important motivation for the use of nanomaterials and nanoarchitectures in cancer therapy emanates from the widespread emergence of drug resistance. Although doxorubicin (DOX) induces cell cycle arrest and DNA damage by suppressing topoisomerase activity, resistance to DOX has severely restricted its anti-cancer potential. Hyaluronic acid (HA) has been extensively utilized for synthesizing nanoparticles as it interacts with CD44 expressed on the surface of cancer cells. Cancer cells can take up HA-modified nanoparticles through receptor-mediated endocytosis. Various types of nanostructures such as carbon nanomaterials, lipid nanoparticles and polymeric nanocarriers have been modified with HA to enhance the delivery of DOX to cancer cells. Hyaluronic acid-based advanced materials provide a platform for the co-delivery of genes and drugs along with DOX to enhance the efficacy of anti-cancer therapy and overcome chemoresistance. In the present review, the potential methods and application of HA-modified nanostructures for DOX delivery in anti-cancer therapy are discussed.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956, Istanbul, Turkey; Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | | | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Nicolas H Voelcker
- Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia; Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria, 3168, Australia; Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Department of Translational Sciences, Xsphera Biosciences Inc., Boston, MA, USA
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Radiobiology Research Center, Iran University of Medical Science, Tehran, Iran
| | - Rajender S Varma
- Regional Center of Advanced Technologies and Materials, Palacky University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - I J Arostegi
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - M Alzola
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, Edinburgh EH9 3JG, UK; Department of Mechanical Engineering, School of Engineering, Shiv Nadar University, Uttar Pradesh 201314, India
| | - Noushin Nabavi
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Center for Materials Interfaces, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy.
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, USA.
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain; University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain; Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore.
| |
Collapse
|
27
|
Della Sala F, Fabozzi A, di Gennaro M, Nuzzo S, Makvandi P, Solimando N, Pagliuca M, Borzacchiello A. Advances in Hyaluronic-Acid-Based (Nano)Devices for Cancer Therapy. Macromol Biosci 2021; 22:e2100304. [PMID: 34657388 DOI: 10.1002/mabi.202100304] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the main cause of fatality all over the world with a considerable growth rate. Many biologically active nanoplatforms are exploited for tumor treatment. Of nanodevices, hyaluronic acid (HA)-based systems have shown to be promising candidates for cancer therapy due to their high biocompatibility and cell internalization. Herein, surface functionalization of different nanoparticles (NPs), e.g., organic- and inorganic-based NPs, is highlighted. Subsequently, HA-based nanostructures and their applications in cancer therapy are presented.
Collapse
Affiliation(s)
- Francesca Della Sala
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Antonio Fabozzi
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Mario di Gennaro
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Stefano Nuzzo
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Pooyan Makvandi
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Nicola Solimando
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Maurizio Pagliuca
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| |
Collapse
|
28
|
Li L, Zhang P, Li C, Guo Y, Sun K. In vitro/vivo antitumor study of modified-chitosan/carboxymethyl chitosan "boosted" charge-reversal nanoformulation. Carbohydr Polym 2021; 269:118268. [PMID: 34294300 DOI: 10.1016/j.carbpol.2021.118268] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/12/2021] [Accepted: 05/26/2021] [Indexed: 12/31/2022]
Abstract
Major obstacles in the development of nanoformulations as efficient drug delivery systems are the rapid clearance from blood circulation and lysosomal entrapment. To overcome these problems, a polysaccharide-based core-shell type charge-switchable nanoformulation (CS-LA-DMMA/CMCS/PAMAM@DOX) is constructed to improve antitumor efficacy of DOX. By applying carboxymethyl chitosan (CMCS) as bridge polymer and negatively charged chitosan-derivative as outer shell, the stability and pH-sensitivity of this nanoformulation is promisingly enhanced. Furthermore, the positively charged PAMAM@DOX could escape from lysosomes via "proton sponge effect" and "cationic-anionic interaction with lysosome membranes". Admirable cellular uptake and high apoptosis/necrosis rate were detected in this study. In vitro assays demonstrate that the CS-LA-DMMA/CMCS/PAMAM@DOX was internalized into HepG2 cells predominantly via the clathrin-mediated endocytosis pathway. Excitingly, in vivo studies showed that high accumulation of CS-LA-DMMA/CMCS/PAMAM@DOX in tumor tissue led to enhanced tumor inhibition. Compared with free DOX, the tumor inhibition rate of nanoformulation was improved up to 226%.
Collapse
Affiliation(s)
- Lin 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, PR China
| | - Peng Zhang
- 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, PR China.
| | - Congcong 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, PR China
| | - Yan Guo
- Department of Development Planning & Discipline Construction, Yantai University, Yantai 264005, PR 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, PR China; State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co., Ltd, Yantai 264003, PR China.
| |
Collapse
|
29
|
Yang HY, Meng Du J, Jang MS, Mo XW, Sun XS, Lee DS, Lee JH, Fu Y. CD44-Targeted and Enzyme-Responsive Photo-Cross-Linked Nanogels with Enhanced Stability for In Vivo Protein Delivery. Biomacromolecules 2021; 22:3590-3600. [PMID: 34286578 DOI: 10.1021/acs.biomac.1c00653] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
One of the biggest challenges of the protein delivery system is to realize stable and high protein encapsulation efficiency in blood circulation and rapid release of protein in the targeted tumor cells. To overcome these hurdles, we fabricated enzyme-responsive photo-cross-linked nanogels (EPNGs) through UV-triggered chemical cross-linking of cinnamyloxy groups in the side chain of PEGylation hyaluronic acid (HA) for CD44-targeted transport of cytochrome c (CC). The EPNGs showed high loading efficiency and excellent stability in different biological media. Notably, CC leakage effectively suppressed under physiological conditions but accelerated release in the presence of hyaluronidase, an overexpressed enzyme in tumor cells. Moreover, thiazolylblue tetrazolium bromide (MTT) results indicated that the vacant EPNGs showed excellent nontoxicity, while CC-loaded EPNGs exhibited higher killing efficiency to CD44-positive A549 cells than to CD44-negative HepG2 cells and free CC. Confocal images confirmed that CC-loaded EPNGs could effectively be internalized by CD44-mediated endocytosis pathway and rapidly escape from the endo/lysosomal compartment. Human lung tumor-bearing mice imaging assays further revealed that CC-loaded EPNGs actively target tumor locations. Remarkably, CC-loaded EPNGs also exhibited enhanced antitumor activity with negligible systemic toxicity. These results implied that these EPNGs have appeared as stable and promising nanocarriers for tumor-targeting protein delivery.
Collapse
Affiliation(s)
- Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, P. R. China
| | - Jia Meng Du
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, P. R. China
| | - Moon-Sun Jang
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Xin Wang Mo
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, P. R. China
| | - Xin Shun Sun
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, P. R. China
| | - Doo Sung Lee
- Theranostic Macromolecules Research Center and School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, Republic of Korea
| | - Jung Hee Lee
- Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine and Center for Molecular and Cellular Imaging, Samsung Biomedical Research Institute, Seoul 06351, Republic of Korea
| | - Yan Fu
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, P. R. China
| |
Collapse
|
30
|
Progress in the study of D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) reversing multidrug resistance. Colloids Surf B Biointerfaces 2021; 205:111914. [PMID: 34130211 DOI: 10.1016/j.colsurfb.2021.111914] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 05/28/2021] [Accepted: 06/06/2021] [Indexed: 12/13/2022]
Abstract
Currently, multidrug resistance (MDR) is one of the major reasons for failure in clinical cancer chemotherapy. Overexpression of the ATP binding cassette (ABC) transporter P-glycoprotein (P-gp), which significantly increases the efflux of anticancer drugs from tumor cells, enhances MDR. In the past few decades, four generations of P-gp inhibitors have appeared. However, they are limited in clinical application due to their severe toxic side effects. As a P-gp inhibitor and carrier for loading chemotherapy agents, TPGS has received increasing attention due to its advantages and unique properties of reversing MDR. TPGS is an amphipathic agent that increases the solubility of most chemotherapy drugs and decreases severe side effects. In addition, TPGS is an excellent carrier with P-gp-inhibiting ability. In this review, we summarize the latest articles on TPGS-based nanodelivery systems to prevent MDR.
Collapse
|
31
|
Development of gold nanorods for cancer treatment. J Inorg Biochem 2021; 220:111458. [PMID: 33857697 DOI: 10.1016/j.jinorgbio.2021.111458] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/01/2021] [Accepted: 04/04/2021] [Indexed: 02/06/2023]
Abstract
There has been growing interest in the application of gold nanorods (GNRs) to tumor therapy due to the unique properties they possess. In the past, GNRs were not used in clinical treatments as they lacked stability in vivo and were characterized by potential toxicity. Despite these issues, the significant potential for utilizing GNRs to conduct safe and effective treatments for tumors cannot be ignored. Therefore, it remains crucial to thoroughly investigate the mechanisms behind the toxicity of GNRs in order to provide the means of overcoming obstacles to its full application in the future. This review presents the toxic effects of GNRs, the factors affecting toxicity and the methods to improve biocompatibility, all of which are presently being studied. Finally, we conclude by briefly discussing the current research status of GNRs and provide additional perspective on the challenges involved along with the course of development for GNRs in the future.
Collapse
|
32
|
Hinchliffe JD, Parassini Madappura A, Syed Mohamed SMD, Roy I. Biomedical Applications of Bacteria-Derived Polymers. Polymers (Basel) 2021; 13:1081. [PMID: 33805506 PMCID: PMC8036740 DOI: 10.3390/polym13071081] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
Plastics have found widespread use in the fields of cosmetic, engineering, and medical sciences due to their wide-ranging mechanical and physical properties, as well as suitability in biomedical applications. However, in the light of the environmental cost of further upscaling current methods of synthesizing many plastics, work has recently focused on the manufacture of these polymers using biological methods (often bacterial fermentation), which brings with them the advantages of both low temperature synthesis and a reduced reliance on potentially toxic and non-eco-friendly compounds. This can be seen as a boon in the biomaterials industry, where there is a need for highly bespoke, biocompatible, processable polymers with unique biological properties, for the regeneration and replacement of a large number of tissue types, following disease. However, barriers still remain to the mass-production of some of these polymers, necessitating new research. This review attempts a critical analysis of the contemporary literature concerning the use of a number of bacteria-derived polymers in the context of biomedical applications, including the biosynthetic pathways and organisms involved, as well as the challenges surrounding their mass production. This review will also consider the unique properties of these bacteria-derived polymers, contributing to bioactivity, including antibacterial properties, oxygen permittivity, and properties pertaining to cell adhesion, proliferation, and differentiation. Finally, the review will select notable examples in literature to indicate future directions, should the aforementioned barriers be addressed, as well as improvements to current bacterial fermentation methods that could help to address these barriers.
Collapse
Affiliation(s)
| | | | | | - Ipsita Roy
- Department of Materials Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield S1 3JD, UK; (J.D.H.); (A.P.M.); (S.M.D.S.M.)
| |
Collapse
|
33
|
Hu X, Zhang J, Deng L, Hu H, Hu J, Zheng G. Galactose-Modified PH-Sensitive Niosomes for Controlled Release and Hepatocellular Carcinoma Target Delivery of Tanshinone IIA. AAPS PharmSciTech 2021; 22:96. [PMID: 33694067 PMCID: PMC7946689 DOI: 10.1208/s12249-021-01973-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 02/21/2021] [Indexed: 12/11/2022] Open
Abstract
Increasing the drug tumor-specific accumulation and controlling their release is considered one of the most effective ways to increase the efficacy of drugs. Here, we developed a vesicle system that can target hepatoma and release drugs rapidly within tumor cells. This non-ionic surfactant vesicle is biodegradable. Galactosylated stearate has been used to glycosylate the vesicles to achieve liver targeting; replacement of a portion (Chol:CHEMS = 1:1) of cholesterol by cholesteryl hemisuccinate (CHEMS) allows for a rapid release of drugs in an acidic environment. In vitro release experiments confirmed that galactose-modified pH-sensitive niosomes loaded with tanshinone IIA had excellent drug release performance in acid medium. In vitro experiments using ovarian cancer cells (A2780), colon cancer cells (HCT8), and hepatoma cell (Huh7, HepG2) confirmed that the preparation had specific targeting ability to hepatoma cells compared with free drugs, and this ability was dependent on the galactose content. Furthermore, the preparation also had a more substantial inhibitory effect on tumor cells, and subsequent apoptosis assays and cell cycle analyses further confirmed its enhanced anti-tumor effect. Results of pharmacokinetic experiments confirmed that the vesicle system could significantly extend the blood circulation time of tanshinone IIA, and the larger area under the curve indicated that the preparation had a better drug effect. Thus, the results of biodistribution experiments confirmed the in vivo liver targeting ability of this preparation. Niosomes designed in this manner are expected to be a safe and effective drug delivery system for liver cancer therapy.
Collapse
|
34
|
Ye Z, Li T, Qing D, Sun Y, Chen H, Yu Q, Yan C. Structural elucidation and osteogenic activity of a novel heteropolysaccharide from Alhagi pseudalhagi. Int J Biol Macromol 2021; 171:185-197. [PMID: 33412197 DOI: 10.1016/j.ijbiomac.2020.12.189] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 01/17/2023]
Abstract
Alhagi pseudalhagi, commonly known as camel thorn, is used as an indigenous medicinal plant in China. The present study was designed to elucidate the structure of a novel polysaccharide, APP90-2, isolated from Alhagi pseudalhagi and evaluate its osteogenic activity. A homogeneous polysaccharide (APP90-2) was obtained from A. pseudalhagi via DEAE-52 and Sephacryl S-100 columns, with a molecular weight of 5.9 kDa. Monosaccharide, GC-MS, and NMR analyses showed that APP90-2 consisted of α-l-Rhap-(1→, →3)-α-l-Araf-(1→, →5)-α-l-Araf-(1→, →4)-β-d-Xylp-(1→, α-d-Glcp-(1→, →3,5)-α-l-Araf-(1→, →4)-β-d-GlcAp-(1→, →4)-3-OAc-α-d-Glcp-(1→, →3)-α-d-Galp-(1→, →3)-β-d-GalAp-(1→, →4)-α-d-Galp-(1→, →6)-α-d-Manp-(1→, →4,6)-β-d-Galp-(1→, and →3,6)-β-d-Glcp-(1→ with relative molar ratios of 4.1:1.8:6.1:6.7:1.7:1.0:1.5:2.7:2.4:1.1:2.3:2.6:1.4:2.0. Morphological analyses revealed that APP90-2 interacted with Congo-red and had an obvious honeycomb structure. Additionally, APP90-2 significantly promoted proliferation, differentiation, and mineralization of MC3T3-E1 cells, indicating that APP90-2 exhibited pronounced osteogenic activity. Therefore, our findings suggest that A. pseudalhagi may be used as an alternative medicine or health supplement for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Zhenquan Ye
- Clinical Pharmacy of the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Tianyu Li
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Degang Qing
- Xinjiang Institute of Chinese Materia Medica and Ethnodrug, Urumqi 830002, China
| | - Yu Sun
- Xinjiang Institute of Chinese Materia Medica and Ethnodrug, Urumqi 830002, China
| | - Haiyun Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qian Yu
- Clinical Pharmacy of the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chunyan Yan
- Clinical Pharmacy of the First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
35
|
Liu XY, Wang JQ, Ashby CR, Zeng L, Fan YF, Chen ZS. Gold nanoparticles: synthesis, physiochemical properties and therapeutic applications in cancer. Drug Discov Today 2021; 26:1284-1292. [PMID: 33549529 DOI: 10.1016/j.drudis.2021.01.030] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/12/2020] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Gold nanoparticles (AuNPs) have been shown to be useful as carriers of various anticancer drugs as well as diagnosis platforms. In this review, we discuss the synthesis and physiochemical properties of AuNPs. We also highlight the photothermal and photodynamic properties of AuNPs and relevant applications in therapeutic studies. Furthermore, we review the applications of AuNPs in cancer treatment as and their underlying anticancer mechanisms in multiple types of cancer.
Collapse
Affiliation(s)
- Xin-Yu Liu
- School of Clinical Medicine, Weifang Medical University, Weifang 261053, Shandong, China; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Leli Zeng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA; Precision Medicine Center, Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Ying-Fang Fan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA; Department of Hepatobiliary Surgery I, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA.
| |
Collapse
|
36
|
Abstract
Compared to normal tissue, solid tumors exhibit a lower pH value. Such pH gradient can be used to design pH-sensitive nanogels for selective drug delivery. The acid-sensitive elements in the nanogel cause it to swell/degrade rapidly, followed by rapid drug release.
Collapse
Affiliation(s)
- Zhen Li
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- PR. China
| | - Jun Huang
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- PR. China
- The Seventh Affiliated Hospital of Sun Yat-Sen University
| | - Jun Wu
- School of Biomedical Engineering
- Sun Yat-sen University
- Guangzhou
- PR. China
| |
Collapse
|
37
|
Xu H, Niu M, Yuan X, Wu K, Liu A. CD44 as a tumor biomarker and therapeutic target. Exp Hematol Oncol 2020; 9:36. [PMID: 33303029 PMCID: PMC7727191 DOI: 10.1186/s40164-020-00192-0] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
CD44, a complex transmembrane glycoprotein, exists in multiple molecular forms, including the standard isoform CD44s and CD44 variant isoforms. CD44 participates in multiple physiological processes, and aberrant expression and dysregulation of CD44 contribute to tumor initiation and progression. CD44 represents a common biomarker of cancer stem cells, and promotes epithelial-mesenchymal transition. CD44 is involved in the regulation of diverse vital signaling pathways that modulate cancer proliferation, invasion, metastasis and therapy-resistance, and it is also modulated by a variety of molecules in cancer cells. In addition, CD44 can serve as an adverse prognostic marker among cancer population. The pleiotropic roles of CD44 in carcinoma potentially offering new molecular target for therapeutic intervention. Preclinical and clinical trials for evaluating the pharmacokinetics, efficacy and drug-related toxicity of CD44 monoclonal antibody have been carried out among tumors with CD44 expression. In this review, we focus on current data relevant to CD44, and outline CD44 structure, the regulation of CD44, functional properties of CD44 in carcinogenesis and cancer progression as well as the potential CD44-targeting therapy for cancer management.
Collapse
Affiliation(s)
- Hanxiao Xu
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Mengke Niu
- Department of Medical Oncology, The Affiliated Tumor Hospital of Zhengzhou University: Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Xun Yuan
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Medical Oncology, The Affiliated Tumor Hospital of Zhengzhou University: Henan Cancer Hospital, Zhengzhou, 450008, China. .,Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| | - Aiguo Liu
- Department of Pediatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
38
|
Zhang X, Wu Y, Li Z, Wang W, Wu Y, Pan D, Gu Z, Sheng R, Tomás H, Zhang H, Rodrigues J, Gong Q, Luo K. Glycodendron/pyropheophorbide-a (Ppa)-functionalized hyaluronic acid as a nanosystem for tumor photodynamic therapy. Carbohydr Polym 2020; 247:116749. [PMID: 32829865 DOI: 10.1016/j.carbpol.2020.116749] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 02/05/2023]
Abstract
To enhance the drug delivery efficiency of hyaluronic acid (HA), we designed and prepared glycodendron and pyropheophorbide-a (Ppa)-functionalized HA (HA-Ppa-Dendron) as a nanosystem for cancer photodynamic therapy. Linear Ppa-modified HA (HA-Ppa) was also prepared as a control. Cellular uptake of both polymers by MDA-MB-231 cells led to mitochondrial dysfunction and generation of reactive oxygen species under the irradiation of a laser. Compared to the linear polymer, HA-Ppa-Dendron had higher molecular weight, a more compact nanoscale particle size, and a dendritic structure, resulting in a much longer blood circulation time and higher tumor accumulation. HA-Ppa-Dendron outperformed HA-Ppa in inhibiting cell growth, with 60 % of tumors was eradicated under laser irradiation. Tumor growth inhibition (TGI) up to 99.2 % was achieved from HA-Ppa-Dendron, which was much higher than that of HA-Ppa (50.6 %). Therefore, glycodendron-functionalized HAs by integration of HA and dendritic polymers may act as efficient anti-cancer nanomedicine.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; College of Preclinical Medicine, Southwest Medical University, Luzhou, 646000, China
| | - Yahui Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhiqian Li
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjia Wang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaping Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruilong Sheng
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Portugal
| | - Helena Tomás
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Portugal
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, CA 91711, USA
| | - João Rodrigues
- CQM - Centro de Química da Madeira, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Portugal
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
39
|
Zhou X, Zhao W, Wang M, Zhang S, Li Y, Hu W, Ren L, Luo S, Chen Z. Dual-Modal Therapeutic Role of the Lactate Oxidase-Embedded Hierarchical Porous Zeolitic Imidazolate Framework as a Nanocatalyst for Effective Tumor Suppression. ACS APPLIED MATERIALS & INTERFACES 2020; 12:32278-32288. [PMID: 32580547 DOI: 10.1021/acsami.0c05783] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The increasing evidence supports the fact that lactate in the tumor microenvironment (TME) plays a vital role in tumor proliferation, metastasis, and recurrence, which in turn is emerging as one of the most interesting molecular targets for tumor treatment. Here, hierarchical porous zeolitic imidazolate framework-8 (ZIF-8) as the nanocarrier is fabricated to simultaneously load lactate oxidase (LOD) and Fe3O4 nanoparticles (NPs), called LOD & Fe3O4@ZIF-8 NPs (LFZ NPs), for tumor therapy. On one hand, the sharp consumption of lactate in the TME by LOD will change the essential "soil" where tumor cells live so as to suppress tumor rapid growth. On the other hand, hydrogen peroxide (H2O2) is produced in the TME from the oxidation of lactate catalyzed by LOD and subsequently converted to highly toxic hydroxyl radicals (•OH) catalyzed by Fe3O4 NPs via Fenton-like reactions to kill tumor cells. Based on the endogenous catalysis, this dual-modal strategy of tumor therapy based on lactate is simple, safe, and effective, which deserves to be well concerned.
Collapse
Affiliation(s)
- Xi Zhou
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Wen Zhao
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Muxue Wang
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Shuai Zhang
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Yunhong Li
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, School of Electronic Science and Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Wenxin Hu
- Harvard College, Harvard University, 209 Dunster Mail Center, 945 Memorial Drive, Cambridge, Massachusetts 02138, United States
| | - Lei Ren
- Department of Biomaterials, College of Materials, Xiamen University, Xiamen, Fujian 361005, P. R. China
| | - Shenglin Luo
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, P. R. China
| | - Zhiwei Chen
- Department of Electronic Science, Fujian Provincial Key Laboratory of Plasma and Magnetic Resonance Research, School of Electronic Science and Engineering, Xiamen University, Xiamen, Fujian 361005, P. R. China
| |
Collapse
|
40
|
Wang K, Xiang Y, Pan W, Wang H, Li N, Tang B. Dual-targeted photothermal agents for enhanced cancer therapy. Chem Sci 2020; 11:8055-8072. [PMID: 34123080 PMCID: PMC8163445 DOI: 10.1039/d0sc03173a] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 07/15/2020] [Indexed: 12/15/2022] Open
Abstract
Photothermal therapy, in which light is converted into heat and triggers local hyperthermia to ablate tumors, presents an inherently specific and noninvasive treatment for tumor tissues. In this area, the development of efficient photothermal agents (PTAs) has always been a central topic. Although many efforts have been made on the investigation of novel molecular architectures and photothermal materials over the past decades, PTAs can cause severe damage to normal tissues because of the poor tumor aggregate ability and high irradiation density. Recently, dual-targeted photothermal agents (DTPTAs) provide an attractive strategy to overcome these problems and enhance cancer therapy. DTPTAs are functionalized with two classes of targeting units, including tumor environment targeting sites, tumor targeting sites and organelle targeting sites. In this perspective, typical targeted ligands and representative examples of photothermal therapeutic agents with dual-targeted properties are systematically summarized and recent advances using DTPTAs in tumor therapy are highlighted.
Collapse
Affiliation(s)
- Kaiye Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Yanan Xiang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Hongyu Wang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Centre of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University Jinan 250014 P. R. China
| |
Collapse
|
41
|
Wang J, Muhammad N, Li T, Wang H, Liu Y, Liu B, Zhan H. Hyaluronic Acid-Coated Camptothecin Nanocrystals for Targeted Drug Delivery to Enhance Anticancer Efficacy. Mol Pharm 2020; 17:2411-2425. [PMID: 32437163 DOI: 10.1021/acs.molpharmaceut.0c00161] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumor-targeted drug delivery via chemotherapy is very effective on cancer treatment. For potential anticancer agent such as Camptothecin (CPT), high chemotherapeutic efficacy and accurate tumor targeting are equally crucial. Inspired by special CD44 binding capability from hyaluronic acid (HA), in this study, novel HA-coated CPT nanocrystals were successfully prepared by an antisolvent precipitation method for tumor-targeted delivery of hydrophobic drug CPT. These HA-coated CPT nanocrystals demonstrated high drug loading efficiency, improved aqueous dispersion, prolonged circulation, and enhanced stability resulting from their nanoscaled sizes and hydrophilic HA layer. Moreover, as compared to crude CPT and naked CPT nanocrystals, HA-coated CPT nanocrystals displayed dramatically enhanced in vitro anticancer activity, apoptosis-inducing potency against CD44 overexpressed cancer cells, and lower toxic effect toward normal cells due to pH-responsive drug release behavior and specific HA-CD44 mediated endocytosis. Additionally, HA-coated CPT nanocrystals performed fairly better antimigration activity and biocompatibility. The possible molecular mechanism regarding this novel drug formulation might be linked to intrinsic mitochondria-mediated apoptosis by an increase of Bax to Bcl-2 ratio and upregulation of P53. Consequently, HA-coated CPT nanocrystals are expected to be an effective nanoplatform in drug delivery for cancer therapy.
Collapse
Affiliation(s)
- Jihui Wang
- Department of Biotechnology, School of Bioengineering, Dalian Polytechnic University, Dalian 116034, Liaoning Province, P. R. China.,School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan 523808, Guangzhou Province, P. R. China
| | - Nazim Muhammad
- Department of Biotechnology, School of Bioengineering, Dalian Polytechnic University, Dalian 116034, Liaoning Province, P. R. China
| | - Tongtong Li
- Department of Biotechnology, School of Bioengineering, Dalian Polytechnic University, Dalian 116034, Liaoning Province, P. R. China
| | - Han Wang
- Department of Biotechnology, School of Bioengineering, Dalian Polytechnic University, Dalian 116034, Liaoning Province, P. R. China
| | - Yujia Liu
- Department of Biotechnology, School of Bioengineering, Dalian Polytechnic University, Dalian 116034, Liaoning Province, P. R. China
| | - Bingnan Liu
- Department of Biotechnology, School of Bioengineering, Dalian Polytechnic University, Dalian 116034, Liaoning Province, P. R. China
| | - Honglei Zhan
- Department of Biotechnology, School of Bioengineering, Dalian Polytechnic University, Dalian 116034, Liaoning Province, P. R. China
| |
Collapse
|
42
|
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
|
43
|
Yang HY, Li Y, Jang MS, Fu Y, Wu T, Lee JH, Lee DS. Green preparation of pH-responsive and dual targeting hyaluronic acid nanogels for efficient protein delivery. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.109342] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|