1
|
Lorenc P, Dams-Kozlowska H, Guzniczak N, Florczak-Substyk A. Application of nanoparticles to target tumor blood vessels as a promising cancer treatment strategy. Biomed Pharmacother 2025; 186:118038. [PMID: 40215646 DOI: 10.1016/j.biopha.2025.118038] [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: 02/10/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
Cancer remains one of the leading causes of death worldwide and poses a significant challenge to effective treatment due to its complexity. Angiogenesis, the formation of new blood vessels, is a critical process in tumor growth and metastasis. The VEGF/VEGFR pathway plays a crucial role in regulating angiogenesis. Many anti-angiogenesis agents, including monoclonal antibodies and tyrosine kinase inhibitors, have been investigated for the treatment of various cancers. However, they face significant limitations such as limited bioavailability and drug resistance. Nanoparticles have emerged as a promising tool for effective drug delivery while minimizing systemic side effects. This review explores the application of nanoparticles dedicated to angiogenesis-targeted cancer therapy, particularly targeting the VEGF/VEGFR pathway. We describe drug delivery systems based on inorganic, lipid, and polymeric nanoparticles. Moreover, special attention is given to functionalized nanoparticles, which can precisely target numerous proteins that are significantly overexpressed on the surfaces of endothelial cells, tumors, or other cells in the tumor microenvironment. We summarize a series of nanoparticles designed for selective targeting of tumor vasculature, emphasizing the challenges faced by anti-angiogenic cancer therapies.
Collapse
Affiliation(s)
- Patryk Lorenc
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61-866, Poland; Doctoral School, Poznan University of Medical Sciences, 70 Bukowska St, Poznan 60-812, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61-866, Poland
| | - Natalia Guzniczak
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland
| | - Anna Florczak-Substyk
- Chair of Medical Biotechnology, Department of Cancer Immunology, Poznan University of Medical Sciences, 8 Rokietnicka St, Poznan 60-806, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, Poznan 61-866, Poland.
| |
Collapse
|
2
|
Cai Y, Chai T, Nguyen W, Liu J, Xiao E, Ran X, Ran Y, Du D, Chen W, Chen X. Phototherapy in cancer treatment: strategies and challenges. Signal Transduct Target Ther 2025; 10:115. [PMID: 40169560 PMCID: PMC11961771 DOI: 10.1038/s41392-025-02140-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/11/2024] [Accepted: 01/13/2025] [Indexed: 04/03/2025] Open
Abstract
Phototherapy has emerged as a promising modality in cancer treatment, garnering considerable attention for its minimal side effects, exceptional spatial selectivity, and optimal preservation of normal tissue function. This innovative approach primarily encompasses three distinct paradigms: Photodynamic Therapy (PDT), Photothermal Therapy (PTT), and Photoimmunotherapy (PIT). Each of these modalities exerts its antitumor effects through unique mechanisms-specifically, the generation of reactive oxygen species (ROS), heat, and immune responses, respectively. However, significant challenges impede the advancement and clinical application of phototherapy. These include inadequate ROS production rates, subpar photothermal conversion efficiency, difficulties in tumor targeting, and unfavorable physicochemical properties inherent to traditional phototherapeutic agents (PTs). Additionally, the hypoxic microenvironment typical of tumors complicates therapeutic efficacy due to limited agent penetration in deep-seated lesions. To address these limitations, ongoing research is fervently exploring innovative solutions. The unique advantages offered by nano-PTs and nanocarrier systems aim to enhance traditional approaches' effectiveness. Strategies such as generating oxygen in situ within tumors or inhibiting mitochondrial respiration while targeting the HIF-1α pathway may alleviate tumor hypoxia. Moreover, utilizing self-luminescent materials, near-infrared excitation sources, non-photoactivated sensitizers, and wireless light delivery systems can improve light penetration. Furthermore, integrating immunoadjuvants and modulating immunosuppressive cell populations while deploying immune checkpoint inhibitors holds promise for enhancing immunogenic cell death through PIT. This review seeks to elucidate the fundamental principles and biological implications of phototherapy while discussing dominant mechanisms and advanced strategies designed to overcome existing challenges-ultimately illuminating pathways for future research aimed at amplifying this intervention's therapeutic efficacy.
Collapse
Affiliation(s)
- Yeyu Cai
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Tian Chai
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, Shanxi Province, China
| | - William Nguyen
- School of Chips, XJTLU Entrepreneur College (Taicang), Xi'an Jiaotong-Liverpool University, Taicang, Suzhou, China
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Enhua Xiao
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China
| | - Xin Ran
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuping Ran
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dan Du
- Department of Dermatovenereology, The West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Chen
- Department of Radiology, Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), Xi'an, Shanxi Province, China.
| | - Xiangyu Chen
- Department of Radiology, The Second Xiangya Hospital of Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
3
|
Naskar A, Kilari S, Baranwal G, Kane J, Misra S. Nanoparticle-Based Drug Delivery for Vascular Applications. Bioengineering (Basel) 2024; 11:1222. [PMID: 39768040 PMCID: PMC11673055 DOI: 10.3390/bioengineering11121222] [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: 08/07/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 01/05/2025] Open
Abstract
Nanoparticle (NP)-based drug delivery systems have received widespread attention due to the excellent physicochemical properties of nanomaterials. Different types of NPs such as lipid NPs, poly(lactic-co-glycolic) acid (PLGA) NPs, inorganic NPs (e.g., iron oxide and Au), carbon NPs (graphene and carbon nanodots), 2D nanomaterials, and biomimetic NPs have found favor as drug delivery vehicles. In this review, we discuss the different types of customized NPs for intravascular drug delivery, nanoparticle behaviors (margination, adhesion, and endothelium uptake) in blood vessels, and nanomaterial compatibility for successful drug delivery. Additionally, cell surface protein targets play an important role in targeted drug delivery, and various vascular drug delivery studies using nanoparticles conjugated to these proteins are reviewed. Finally, limitations, challenges, and potential solutions for translational research regarding NP-based vascular drug delivery are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (A.N.); (S.K.); (G.B.); (J.K.)
| |
Collapse
|
4
|
Wu H, Jin M, Liu Y, Wang S, Liu C, Quan X, Jin M, Gao Z, Jin Y. A self-targeting MOFs nanoplatform for treating metastatic triple-negative breast cancer through tumor microenvironment remodeling and chemotherapy potentiation. Int J Pharm 2024; 664:124625. [PMID: 39182743 DOI: 10.1016/j.ijpharm.2024.124625] [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: 05/06/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and fatal subtype of breast cancer with disappointing treatment and high mortality. Tumor microenvironment (TME) plays an important role in the invasion and metastasis of TNBC through multiple complex processes. Most anti-metastatic therapies only focus on cancer cells themselves or interfering with single factors of the metastasis process, which is often related to poor outcomes. Thus, effective TNBC treatment relies on regulating multiple key metastasis-related aspects of the TME. Herein, a self-targeting Metal-Organic Frameworks (MOFs) nanoplatform (named as MTX-PEG@TPL@ZIF-8) was designed to improve treatment of TNBC through tumor microenvironment remodeling and chemotherapy potentiation. The self-targeting MOF nanoplatform is consist of ZIF-8 nanoparticles loaded triptolide (TPL) and followed by the coating with methotrexate-polyethylene glycol conjugates (MTX-PEG). Due to MTX's affinity for the overexpressed folate receptor on tumor cell surfaces, MTX-PEG@TPL@ZIF-8 enables effective accumulation and deep penetration in the tumor area by an MTX-mediated self-targeting strategy. This MOF nanoplatform could promptly release the medication after penetrating the tumor cell, due to pH-triggered degradation. Its anti-metastasis mechanism is to inhibit tumor invasion and metastasis by down-regulating the expression of Vimentin, MMP-2 and MMP-9 and increasing the expression of E-cadherin, upregulation of cleaved caspase-3 and cleaved caspase-9 protein expression promote the apoptosis of tumor cells, thereby reducing their migration. It also downregulated the expression of VEGF and CD31 protein to inhibit the generation of neovascularization. Overall, these findings suggest the self-targeting MOF nanoplatform offers new insights into the treatment of metastatic TNBC by TME remodeling and potentiating chemotherapy.
Collapse
Affiliation(s)
- Hao Wu
- Department of Pharmacy, Jilin Medical University, Jilin, Jilin Province 132013, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ming Jin
- Department of Orthopedic Surgery, Yanbian University Hospital, Yanji, Jilin Province 133000, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shuangqing Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Chao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiuquan Quan
- Department of Orthopedic Surgery, Yanbian University Hospital, Yanji, Jilin Province 133000, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Ying Jin
- Department of Pharmacy, Jilin Medical University, Jilin, Jilin Province 132013, China.
| |
Collapse
|
5
|
El-Sofany WI, Azzam EMS, Latif S, Hamden K. Spirothiazolidine-Derivative on Silver Nanoparticles and Carbon Nanotubes: Evaluation of Antibacterial, Anti-Fungal, Anti-Inflammatory, Antioxidant and Gastroprotective Activities. Pharmaceutics 2024; 16:901. [PMID: 39065599 PMCID: PMC11279549 DOI: 10.3390/pharmaceutics16070901] [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: 05/08/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
This study aims to develop innovative heterocyclic nanocomposites incorporating silver nanoparticles (SNPs) for potential therapeutic applications targeting infections, gastric ulceration, inflammation, and oxidative damage. By synthesizing new derivatives of spiro-thiazolidine-carbonitrile (Py-ST-X) and incorporating them into Ag nanoparticles (AgNPs) and carbon nanotubes (CNTs), we have prepared Ag@Py-ST-X and Ag@Py-ST-X@CNT nanocomposites, respectively. The physical properties of these materials were studied using XRD, TEM, SEM, and Zeta potential techniques. In our investigation involving rats with gastric ulcers, we observed noteworthy inhibitory effects on gastric acid enzyme activity, specifically H+/K+ATPase, by Ag@Py-ST-NO2 and Ag@Py-ST-Br nanocomposites, demonstrating reductions of 25 and 34%, respectively, compared to untreated ulcers. Nanotubulation of these compounds further improved their inhibitory efficacy to 29 and 45%, respectively. Additionally, these nanoparticles showed the most potent myeloperoxidase (MPO)-inhibitory activity, demonstrating 36 and 49% inhibition, respectively, with nanotubulated versions reaching 44 and 53%. Moreover, Ag@Py-ST-NO2@CNT and Ag@Py-ST-Br@CNT nanotubes showed significant antioxidant activity, reducing thiobarbituric acid reactive substances (TBARS) by 35 and 51%, and hydrogen peroxide (H2O2) levels by 49 and 71%, respectively. These therapeutic effects were confirmed by reductions in gastric surface area (GSA) by 44% and 52%, a decrease in ulcer index (UI) from 80% to 44 and 38%, and an increase in curative index (CI) from 19 to 55 and 62% following administration of Ag@Py-ST-NO2@CNT and Ag@Py-ST-Br@CNT, respectively. Histological studies support these findings, suggesting the potential of these nanocomposites as promising candidates for treating various disorders.
Collapse
Affiliation(s)
- Walaa I. El-Sofany
- Department of Chemistry, College of Science, University of Ha’il, Ha’il 81451, Saudi Arabia
- Photochemistry Department, Chemical Industries Research Institute, National Research Centre, 33 EL Buhouth St., Dokki, Giza 12622, Egypt
| | - Eid. M. S. Azzam
- Department of Chemistry, College of Science, University of Ha’il, Ha’il 81451, Saudi Arabia
- Egyptian Petroleum Research Institute, Nasr City, Cairo 11727, Egypt
| | - Salman Latif
- Department of Chemistry, College of Science, University of Ha’il, Ha’il 81451, Saudi Arabia
| | - Khaled Hamden
- Laboratory of Bioresources: Integrative Biology and Exploiting, Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir 5000, Tunisia
| |
Collapse
|
6
|
Yazdani S, Mozaffarian M, Pazuki G, Hadidi N, Villate-Beitia I, Zárate J, Puras G, Pedraz JL. Carbon-Based Nanostructures as Emerging Materials for Gene Delivery Applications. Pharmaceutics 2024; 16:288. [PMID: 38399344 PMCID: PMC10891563 DOI: 10.3390/pharmaceutics16020288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/03/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Gene therapeutics are promising for treating diseases at the genetic level, with some already validated for clinical use. Recently, nanostructures have emerged for the targeted delivery of genetic material. Nanomaterials, exhibiting advantageous properties such as a high surface-to-volume ratio, biocompatibility, facile functionalization, substantial loading capacity, and tunable physicochemical characteristics, are recognized as non-viral vectors in gene therapy applications. Despite progress, current non-viral vectors exhibit notably low gene delivery efficiency. Progress in nanotechnology is essential to overcome extracellular and intracellular barriers in gene delivery. Specific nanostructures such as carbon nanotubes (CNTs), carbon quantum dots (CQDs), nanodiamonds (NDs), and similar carbon-based structures can accommodate diverse genetic materials such as plasmid DNA (pDNA), messenger RNA (mRNA), small interference RNA (siRNA), micro RNA (miRNA), and antisense oligonucleotides (AONs). To address challenges such as high toxicity and low transfection efficiency, advancements in the features of carbon-based nanostructures (CBNs) are imperative. This overview delves into three types of CBNs employed as vectors in drug/gene delivery systems, encompassing their synthesis methods, properties, and biomedical applications. Ultimately, we present insights into the opportunities and challenges within the captivating realm of gene delivery using CBNs.
Collapse
Affiliation(s)
- Sara Yazdani
- Department of Chemical Engineering, Amirkabir University of Technology, Tehran P.O. Box 15875-4413, Iran; (S.Y.); (G.P.)
- NanoBioCel Research Group, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (I.V.-B.); (J.Z.); (G.P.)
| | - Mehrdad Mozaffarian
- Department of Chemical Engineering, Amirkabir University of Technology, Tehran P.O. Box 15875-4413, Iran; (S.Y.); (G.P.)
| | - Gholamreza Pazuki
- Department of Chemical Engineering, Amirkabir University of Technology, Tehran P.O. Box 15875-4413, Iran; (S.Y.); (G.P.)
| | - Naghmeh Hadidi
- Department of Clinical Research and EM Microscope, Pasteur Institute of Iran (PII), Tehran P.O. Box 131694-3551, Iran;
| | - Ilia Villate-Beitia
- NanoBioCel Research Group, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (I.V.-B.); (J.Z.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain
| | - Jon Zárate
- NanoBioCel Research Group, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (I.V.-B.); (J.Z.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain
| | - Gustavo Puras
- NanoBioCel Research Group, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (I.V.-B.); (J.Z.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain
| | - Jose Luis Pedraz
- NanoBioCel Research Group, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (I.V.-B.); (J.Z.); (G.P.)
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Av Monforte de Lemos 3-5, 28029 Madrid, Spain
- Bioaraba, NanoBioCel Research Group, Calle José Achotegui s/n, 01009 Vitoria-Gasteiz, Spain
| |
Collapse
|
7
|
Yeo M, Sarkar A, Singh YP, Derman ID, Datta P, Ozbolat IT. Synergistic coupling between 3D bioprinting and vascularization strategies. Biofabrication 2023; 16:012003. [PMID: 37944186 PMCID: PMC10658349 DOI: 10.1088/1758-5090/ad0b3f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 09/27/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023]
Abstract
Three-dimensional (3D) bioprinting offers promising solutions to the complex challenge of vascularization in biofabrication, thereby enhancing the prospects for clinical translation of engineered tissues and organs. While existing reviews have touched upon 3D bioprinting in vascularized tissue contexts, the current review offers a more holistic perspective, encompassing recent technical advancements and spanning the entire multistage bioprinting process, with a particular emphasis on vascularization. The synergy between 3D bioprinting and vascularization strategies is crucial, as 3D bioprinting can enable the creation of personalized, tissue-specific vascular network while the vascularization enhances tissue viability and function. The review starts by providing a comprehensive overview of the entire bioprinting process, spanning from pre-bioprinting stages to post-printing processing, including perfusion and maturation. Next, recent advancements in vascularization strategies that can be seamlessly integrated with bioprinting are discussed. Further, tissue-specific examples illustrating how these vascularization approaches are customized for diverse anatomical tissues towards enhancing clinical relevance are discussed. Finally, the underexplored intraoperative bioprinting (IOB) was highlighted, which enables the direct reconstruction of tissues within defect sites, stressing on the possible synergy shaped by combining IOB with vascularization strategies for improved regeneration.
Collapse
Affiliation(s)
- Miji Yeo
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
| | - Anwita Sarkar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Yogendra Pratap Singh
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
| | - Irem Deniz Derman
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, United States of America
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, United States of America
- Department of Biomedical Engineering, Penn State University, University Park, PA 16802, United States of America
- Materials Research Institute, Penn State University, University Park, PA 16802, United States of America
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, United States of America
- Penn State Cancer Institute, Penn State University, Hershey, PA 17033, United States of America
- Biotechnology Research and Application Center, Cukurova University, Adana 01130, Turkey
| |
Collapse
|
8
|
Won Lee J, Kyu Shim M, Kim H, Jang H, Lee Y, Hwa Kim S. RNAi therapies: Expanding applications for extrahepatic diseases and overcoming delivery challenges. Adv Drug Deliv Rev 2023; 201:115073. [PMID: 37657644 DOI: 10.1016/j.addr.2023.115073] [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: 04/27/2023] [Revised: 07/31/2023] [Accepted: 08/20/2023] [Indexed: 09/03/2023]
Abstract
The era of RNA medicine has become a reality with the success of messenger RNA (mRNA) vaccines against COVID-19 and the approval of several RNA interference (RNAi) agents in recent years. Particularly, therapeutics based on RNAi offer the promise of targeting intractable and previously undruggable disease genes. Recent advances have focused in developing delivery systems to enhance the poor cellular uptake and insufficient pharmacokinetic properties of RNAi therapeutics and thereby improve its efficacy and safety. However, such approach has been mainly achieved via lipid nanoparticles (LNPs) or chemical conjugation with N-Acetylgalactosamine (GalNAc), thus current RNAi therapy has been limited to liver diseases, most likely to encounter liver-targeting limitations. Hence, there is a huge unmet medical need for intense evolution of RNAi therapeutics delivery systems to target extrahepatic tissues and ultimately extend their indications for treating various intractable diseases. In this review, challenges of delivering RNAi therapeutics to tumors and major organs are discussed, as well as their transition to clinical trials. This review also highlights innovative and promising preclinical RNAi-based delivery platforms for the treatment of extrahepatic diseases.
Collapse
Affiliation(s)
- Jong Won Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Man Kyu Shim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hyosuk Kim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hochung Jang
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Yuhan Lee
- Department of Anesthesiology, Perioperative, and Pain Medicine, Center for Accelerated Medical Innovation & Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Sun Hwa Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| |
Collapse
|
9
|
Wang Y, Wei Y, Chen L, Yang Y, Jia F, Yu W, Zhou S, Yu S. Research progress of siVEGF complex and their application in antiangiogenic therapy. Int J Pharm 2023; 643:123251. [PMID: 37481098 DOI: 10.1016/j.ijpharm.2023.123251] [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: 02/21/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Vascular endothelial growth factor (VEGF) is an important factor in the development of some diseases such as tumors, ocular neovascular disease and endometriosis. Inhibition of abnormal VEGF expression is one of the most effective means of treating these diseases. The resistance and side effects of currently used VEGF drugs limit their application. Herein, small interfering RNA for VEGF (siVEGF) are developed to inhibit VEGF expression at the genetic level by means of RNA interference. However, as a foreign substance entering the organism, siVEGF is prone to induce an immune response or mismatch, which adversely affects the organism. It is also subjected to enzymatic degradation and cell membrane blockage, which greatly reduces its therapeutic effect. Targeted siVEGF complexes are constructed by nanocarriers to avoid their clearance by the body and precisely target cells, exerting anti-vascular effects for the treatment of relevant diseases. In addition, some multifunctional complexes allow for the combination of siVEGF with other therapeutic tools to improve the treat efficiency of the disease. Therefore, this review describes the construction of the siVEGF complex, its mechanism of action, application in anti-blood therapy, and provides an outlook on its current problems and prospects.
Collapse
Affiliation(s)
- Yan Wang
- Shanxi Medical University, Taiyuan 030001, China
| | - Yingying Wei
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China
| | - Lin Chen
- Key Laboratory of Interface Science and Engineering in Advanced Materials, Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yongzhen Yang
- Key Laboratory of Interface Science and Engineering in Advanced Materials, Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, China
| | - Fan Jia
- Shanxi Medical University, Taiyuan 030001, China
| | - Weiran Yu
- The Affiliated High School of Shanxi University, Taiyuan 030006, China
| | - Shizhao Zhou
- Shanxi Medical University, Taiyuan 030001, China
| | - Shiping Yu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan 030013, China.
| |
Collapse
|
10
|
Li Y, Si R, Wang J, Hai P, Zheng Y, Zhang Q, Pan X, Zhang J. Discovery of novel antibody-drug conjugates bearing tissue protease specific linker with both anti-angiogenic and strong cytotoxic effects. Bioorg Chem 2023; 137:106575. [PMID: 37148706 DOI: 10.1016/j.bioorg.2023.106575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 05/08/2023]
Abstract
Bevacizumab is an FDA-approved class of monoclonal antibodies used to inhibit angiogenesis and promote normalization of blood vessels. It is usually combined with chemotherapeutic agents to treat a variety of solid tumors. However, the whole-body toxicities and toxicity associated with chemotherapy greatly limit the clinical use of this combination therapy. Antibody-drug conjugates (ADCs) couple monoclonal antibodies to cytotoxic molecules via a linker, utilizing the high specificity of monoclonal antibodies to tumor surface antigens to act as a "biological missile" to deliver chemotherapeutic drugs to the tumor site. Herein, we designed a bevacizumab-based ADC, Bevacizumab Vedotin, conjugating bevacizumab to the microtubulin inhibitor MMAE via a tissue protease-specific linker. Biological studies showed strong stability and good tumor cell targeting of our constructed ADCs; rapid drug release was achieved in the presence of exogenous histone protease B. In addition, Bevacizumab Vedotin exhibited good anti-proliferative, apoptosis-promoting and cell cycle-stalling effects on glioma (U87), hepatocellular carcinoma (HepG2), and breast cancer (MCF-7) cell lines. Further in vitro assays demonstrated the enhanced anti-migration activity against MCF-7, potent anti-angiogenic effects, and blockade of the VEGF/VEGFR pathway of Bevacizumab Vedotin.
Collapse
Affiliation(s)
- Yanchen Li
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ru Si
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Ping Hai
- NMPA Key Laboratory for Quality Control of Traditional Chinese and Tibetan Medicine, Qinghai Provincial Drug Inspection and Testing Institute, Xining 810016, China
| | - Yongbiao Zheng
- NMPA Key Laboratory for Quality Control of Traditional Chinese and Tibetan Medicine, Qinghai Provincial Drug Inspection and Testing Institute, Xining 810016, China
| | - Qingqing Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
11
|
Yadav DN, Ali MS, Thanekar AM, Pogu SV, Rengan AK. Recent Advancements in the Design of Nanodelivery Systems of siRNA for Cancer Therapy. Mol Pharm 2022; 19:4506-4526. [PMID: 36409653 DOI: 10.1021/acs.molpharmaceut.2c00811] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
RNA interference (RNAi) has increased the possibility of restoring RNA drug targets for cancer treatment. Small interfering RNA (siRNA) is a promising therapeutic RNAi tool that targets the defective gene by inhibiting its mRNA expression and stopping its translation. However, siRNAs have flaws like poor intracellular trafficking, RNase degradation, rapid kidney filtration, off-targeting, and toxicity, which limit their therapeutic efficiency. Nanocarriers (NCs) have been designed to overcome such flaws and increase antitumor activity. Combining siRNA and anticancer drugs can give synergistic effects in cancer cells, making them a significant gene-modification tool in cancer therapy. Our discussion of NCs-mediated siRNA delivery in this review includes their mechanism, limitations, and advantages in comparison with naked siRNA delivery. We will also discuss organic NCs (polymers and lipids) and inorganic NCs (quantum dots, carbon nanotubes, and gold) that have been reported for extensive delivery of therapeutic siRNA to tumor sites. Finally, we will conclude by discussing the studies based on organic and inorganic NCs-mediated siRNA drug delivery systems conducted in the years 2020 and 2021.
Collapse
Affiliation(s)
- Dokkari Nagalaxmi Yadav
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | - Mohammad Sadik Ali
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | | | - Sunil Venkanna Pogu
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering Indian Institute of Technology Hyderabad, Kandi 502284, India
| |
Collapse
|
12
|
Li SQ, Yang Y, Ye LS. Angiogenesis and immune checkpoint dual blockade: Opportunities and challenges for hepatocellular carcinoma therapy. World J Gastroenterol 2022; 28:6034-6044. [PMID: 36405383 PMCID: PMC9669824 DOI: 10.3748/wjg.v28.i42.6034] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/06/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
The disease burden related to hepatocellular carcinoma (HCC) is increasing. Most HCC patients are diagnosed at the advanced stage and multikinase inhibitors have been the only treatment choice for them. Recently, the approval of immune checkpoint inhibitors (ICIs) has provided a new therapeutic strategy for HCC. It is noteworthy that the positive outcomes of the phase III clinical trial IMBrave150 [atezolizumab (anti-programmed cell death ligand 1 antibody) combined with bevacizumab (anti-vascular endothelial growth factor monoclonal antibody)], showed that overall survival and progression-free survival were significantly better with sorafenib. This combination therapy has become the new standard therapy for advanced HCC and has also attracted more attention in the treatment of HCC with anti-angiogenesis-immune combination therapy. Currently, the synergistic antitumor efficacy of this combination has been shown in many preclinical and clinical studies. In this review, we discuss the mechanism and clinical application of anti-angiogenics and immunotherapy in HCC, outline the relevant mechanism and rationality of the combined application of anti-angiogenics and ICIs, and point out the existing challenges of the combination therapy.
Collapse
Affiliation(s)
- Si-Qi Li
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | - Yang Yang
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| | - Lin-Sen Ye
- Department of Hepatic Surgery and Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, Guangdong Province, China
| |
Collapse
|
13
|
Jin M, Zeng B, Liu Y, Jin L, Hou Y, Liu C, Liu W, Wu H, Chen L, Gao Z, Huang W. Co-Delivery of Repurposing Itraconazole and VEGF siRNA by Composite Nanoparticulate System for Collaborative Anti-Angiogenesis and Anti-Tumor Efficacy against Breast Cancer. Pharmaceutics 2022; 14:pharmaceutics14071369. [PMID: 35890264 PMCID: PMC9317122 DOI: 10.3390/pharmaceutics14071369] [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: 05/12/2022] [Revised: 06/16/2022] [Accepted: 06/24/2022] [Indexed: 12/04/2022] Open
Abstract
Combinations of two different therapeutic modalities of VEGF inhibitors against angiogenesis can cooperatively impede breast cancer tumor growth and enhance therapeutic efficacy. Itraconazole (ITZ) is a conventional antifungal drug with high safety; however, it has been repurposed to be a multi target anti-angiogenesis agent for cancer therapy in recent years. In the present study, composite nanoparticles co-loaded with ITZ and VEGF siRNA were prepared in order to investigate their anti-angiogenesis efficacy and synergistic anticancer effect against breast cancer. The nanoparticles had a suitable particle size (117.9 ± 10.3 nm) and weak positive surface charge (6.69 ± 2.46 mV), as well as good stability and drug release profile in vitro. Moreover, the nanoparticles successfully escaped from endosomes and realized cell apoptosis and cell proliferation inhibition in vitro. In vitro and in vivo experiments showed that the nanoparticles could induce the silencing of VEGF-related expressions as well as anti-angiogenesis efficacy, and the co-loaded ITZ-VEGF siRNA NPs could inhibit tumor growth effectively with low toxicity and side effects. Taken together, the as-prepared delivery vehicles are a simple and safe nano-platform that improves the antitumor efficacy of VEGF siRNA and ITZ, which allows the repositioning of the generic drug ITZ as a great candidate for antitumor therapy.
Collapse
Affiliation(s)
- Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Bowen Zeng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Respiratory Medicine, Yanbian University Hospital, Yanji 133000, China
| | - Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lili Jin
- Department of Pharmacy, Yanbian University, Yanji 133000, China;
| | - Yan Hou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Pharmacy, Yanbian University, Yanji 133000, China;
| | - Chao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Hao Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Pharmacy, Yanbian University, Yanji 133000, China;
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (Z.G.); (W.H.)
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (M.J.); (B.Z.); (Y.L.); (Y.H.); (C.L.); (W.L.); (H.W.); (L.C.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: (Z.G.); (W.H.)
| |
Collapse
|
14
|
Marin D, Marchesan S. Carbon Graphitization: Towards Greener Alternatives to Develop Nanomaterials for Targeted Drug Delivery. Biomedicines 2022; 10:1320. [PMID: 35740342 PMCID: PMC9220131 DOI: 10.3390/biomedicines10061320] [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: 05/22/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/16/2022] Open
Abstract
Carbon nanomaterials have attracted great interest for their unique physico-chemical properties for various applications, including medicine and, in particular, drug delivery, to solve the most challenging unmet clinical needs. Graphitization is a process that has become very popular for their production or modification. However, traditional conditions are energy-demanding; thus, recent efforts have been devoted to the development of greener routes that require lower temperatures or that use waste or byproducts as a carbon source in order to be more sustainable. In this concise review, we analyze the progress made in the last five years in this area, as well as in their development as drug delivery agents, focusing on active targeting, and conclude with a perspective on the future of the field.
Collapse
Affiliation(s)
| | - Silvia Marchesan
- Chemical and Pharmaceutical Sciences Department, University of Trieste, 34127 Trieste, Italy;
| |
Collapse
|
15
|
Rules of Chinese Herbal Intervention of Radiation Pneumonia Based on Network Pharmacology and Data Mining. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7313864. [PMID: 35509624 PMCID: PMC9060976 DOI: 10.1155/2022/7313864] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 02/11/2022] [Accepted: 04/02/2022] [Indexed: 12/30/2022]
Abstract
Objective To explore the mechanism and principles of traditional Chinese medicine (TCM) in the management of radiation pneumonia. Methods The targets of radiation pneumonia were obtained by screening the GeneCards, OMIM, TTD, DrugBank, and HERB databases, analyzing ADME parameters. In addition, compounds and Chinese herbs that can act on the targets were screened from the TCMSP database. The core target compounds for TCM were used to construct the target-compound, compound-traditional Chinese medicine, and target-compound-traditional Chinese medicine networks. These networks were further used to select the core targets, compounds, and TCM. The binding strength between the core targets and compounds was determined using AutoDock Vina. The trajectory for the molecular dynamics simulation was completed by Desmond version 2020. Results A total of 55 active targets in radiation pneumonia were identified. Subsequently, 137 candidate compounds and 469 Chinese herbs were matched. Frequency statistics showed that the Chinese herbs that could interfere with radiation pneumonia were mainly bitter, spicy, and sweet, with both cold and warm properties. Moreover, they mainly belonged to liver and lung channels. The core targets included TNF, IL-6, TGF-β1, and TP53. The most important components were quercetin, resveratrol, and (-)-epigallocatechin-3-gallate. Moreover, the most significant traditional Chinese herbs were Perilla pueraria, ephedra, Lonicerae japonicae, and sea buckthorn. Furthermore, analysis of 222 sets of receptor-ligand docking results suggested that the compounds had good docking activity to their core targets. By combining the docking binding energy, we determined that the chemical compounds had strong binding energy to the targets. Conclusion Using network pharmacology, we explored the potential mechanism of TCM in the treatment of radiation pneumonia. The general rules for application of TCM in the treatment of radiation pneumonia were summarized. This study provides baseline information for future research on the development of TCM for the management of radiation pneumonia.
Collapse
|
16
|
Li Y, Chen Z, Gu L, Duan Z, Pan D, Xu Z, Gong Q, Li Y, Zhu H, Luo K. Anticancer nanomedicines harnessing tumor microenvironmental components. Expert Opin Drug Deliv 2022; 19:337-354. [PMID: 35244503 DOI: 10.1080/17425247.2022.2050211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Small-molecular drugs are extensively used in cancer therapy, while they have issues of nonspecific distribution and consequent side effects. Nanomedicines that incorporate chemotherapeutic drugs have been developed to enhance the therapeutic efficacy of these drugs and reduce their side effects. One of the promising strategies is to prepare nanomedicines by harnessing the unique tumor microenvironment (TME). AREAS COVERED The TME contains numerous cell types that specifically express specific antibodies on the surface including tumor vascular endothelial cells, tumor-associated adipocytes, tumor-associated fibroblasts, tumor-associated immune cells and cancer stem cells. The physicochemical environment is characterized with a low pH, hypoxia, and a high redox potential resulting from tumor-specific metabolism. The intelligent nanomedicines can be categorized into two groups: the first group which is rapidly responsive to extracellular chemical/biological factors in the TME and the second one which actively and/or specifically targets cellular components in the TME. EXPERT OPINION In this paper, we review recent progress of nanomedicines by harnessing the TME and illustrate the principles and advantages of different strategies for designing nanomedicines, which are of great significance for exploring novel nanomedicines or translating current nanomedicines into clinical practice. We will discuss the challenges and prospects of preparing nanomedicines to utilize or alter the TME for achieving effective, safe anticancer treatment.
Collapse
Affiliation(s)
- Yinggang Li
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhonglan Chen
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Gu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhengyu Duan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dayi Pan
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhuping Xu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Youping Li
- Chinese Evidence-Based Medicine Centre, Cochrane China Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongyan Zhu
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Luo
- Laboratory of Stem Cell Biology, Department of Cardiology, Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.,Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
17
|
Yang Z, Deng W, Zhang X, An Y, Liu Y, Yao H, Zhang Z. Opportunities and Challenges of Nanoparticles in Digestive Tumours as Anti-Angiogenic Therapies. Front Oncol 2022; 11:789330. [PMID: 35083147 PMCID: PMC8784389 DOI: 10.3389/fonc.2021.789330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/10/2021] [Indexed: 01/04/2023] Open
Abstract
Digestive tumours, a common kind of malignancy worldwide, have recently led to the most tumour-related deaths. Angiogenesis, the process of forming novel blood vessels from pre-existing vessels, is involved in various physiological and pathological processes in the body. Many studies suggest that abnormal angiogenesis plays an important role in the growth, progression, and metastasis of digestive tumours. Therefore, anti-angiogenic therapy is considered a promising target for improving therapeutic efficacy. Traditional strategies such as bevacizumab and regorafenib can target and block the activity of proangiogenic factors to treat digestive tumours. However, due to resistance and some limitations, such as poor pharmacokinetics, their efficacy is not always satisfactory. In recent years, nanotechnology-based anti-angiogenic therapies have emerged as a new way to treat digestive tumours. Compared with commonly used drugs, nanoparticles show great potential in tumour targeted delivery, controlled drug release, prolonged cycle time, and increased drug bioavailability. Therefore, anti-angiogenic nanoparticles may be an effective complementary therapy to treat digestive tumours. In this review, we outline the different mechanisms of angiogenesis, the effects of nanoparticles on angiogenesis, and their biomedical applications in various kinds of digestive tumours. In addition, the opportunities and challenges are briefly discussed.
Collapse
Affiliation(s)
| | | | | | | | | | - Hongwei Yao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University and National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
18
|
Zhu D, Li Y, Zhang Z, Xue Z, Hua Z, Luo X, Zhao T, Lu C, Liu Y. Recent advances of nanotechnology-based tumor vessel-targeting strategies. J Nanobiotechnology 2021; 19:435. [PMID: 34930293 PMCID: PMC8686559 DOI: 10.1186/s12951-021-01190-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Tumor vessels can provide oxygen and nutrition for solid tumor tissue, create abnormal tumor microenvironment (TME), and play a vital role in the development, immune escape, metastasis and drug resistance of tumor. Tumor vessel-targeting therapy has become an important and promising direction in anti-tumor therapy, with the development of five anti-tumor therapeutic strategies, including vascular disruption, anti-angiogenesis, vascular blockade, vascular normalization and breaking immunosuppressive TME. However, the insufficient drug accumulation and severe side effects of vessel-targeting drugs limit their development in clinical application. Nanotechnology offers an excellent platform with flexible modified surface that can precisely deliver diverse cargoes, optimize efficacy, reduce side effects, and realize the combined therapy. Various nanomedicines (NMs) have been developed to target abnormal tumor vessels and specific TME to achieve more efficient vessel-targeting therapy. The article reviews tumor vascular abnormalities and the resulting abnormal microenvironment, the application of NMs in the tumor vessel-targeting strategies, and how NMs can improve these strategies and achieve multi-strategies combination to maximize anti-tumor effects. ![]()
Collapse
Affiliation(s)
- Dongjie Zhu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhengjia Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zeyu Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhenglai Hua
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xinyi Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ting Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
19
|
Tang Y, Jia C, Wang Y, Wan W, Li H, Huang G, Zhang X. Lactate Consumption via Cascaded Enzymes Combined VEGF siRNA for Synergistic Anti-Proliferation and Anti-Angiogenesis Therapy of Tumors. Adv Healthc Mater 2021; 10:e2100799. [PMID: 34310079 DOI: 10.1002/adhm.202100799] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/08/2021] [Indexed: 02/07/2023]
Abstract
Lactate, as the most abundant component with concentrations of 4-40 mm in tumors, contributes to the regulation of metabolic pathways, angiogenesis, and immunosuppression, exhibiting remarkable potential in cancer treatment. Therefore, a codelivery strategy that combined the cascaded enzymes Lactate oxidase/Catalase (LOx/CAT) and vascular endothelial growth factor (VEGF) siRNA (siVEGF) to suppress tumor proliferation and angiogenesis synergistically is creatively proposed. In brief, the cationic liposomes (LIP) encapsulated with LOx/CAT and siVEGF via hydrophilic interaction and electrostatic adsorption followed by coating with PEGylated phenylboronic acid (PP) is established (PPL@[LOX+CAT]). Moreover, a simple 3-aminophenylboronic acid (PBA)-shielded strategy via fructose (Fru) is applied to further enhance the targeting efficiency in the tumor site. The obtained co-encapsulated nanoparticles (NPs) can simultaneous intracellular release of LOx/CAT and siVEGF, and the collaborative use of LOx and CAT can promote lactate consumption even under a hypoxic tumor microenvironment (TME) without producing systemic toxicity. The combined application of lactate depletion and VEGF silencing demonstrated the efficient migration suppression of 4T1 cells in vitro and superior antitumor and antimetastatic properties in vivo. This work offers a promising tumor treatment strategy via integrating cascaded enzymes and gene therapy, and explores a promising therapy regimen for 4T1 triple-negative breast cancer.
Collapse
Affiliation(s)
- Yan Tang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Changhao Jia
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Yu Wang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Wenjun Wan
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Hui Li
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Gui Huang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| | - Xuenong Zhang
- Department of Pharmaceutics College of Pharmaceutical Sciences Soochow University Suzhou 215123 China
| |
Collapse
|
20
|
Tang L, Mei Y, Shen Y, He S, Xiao Q, Yin Y, Xu Y, Shao J, Wang W, Cai Z. Nanoparticle-Mediated Targeted Drug Delivery to Remodel Tumor Microenvironment for Cancer Therapy. Int J Nanomedicine 2021; 16:5811-5829. [PMID: 34471353 PMCID: PMC8403563 DOI: 10.2147/ijn.s321416] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/14/2021] [Indexed: 12/24/2022] Open
Abstract
Advanced research has revealed the crucial role of tumor microenvironment (TME) in tumorigenesis. TME consists of a complicated network with a variety of cell types including endothelial cells, pericytes, immune cells, cancer-associated fibroblasts (CAFs), cancer stem cells (CSCs) as well as the extracellular matrix (ECM). The TME-constituting cells interact with the cancerous cells through plenty of signaling mechanisms and pathways in a dynamical way, participating in tumor initiation, progression, metastasis, and response to therapies. Hence, TME is becoming an attractive therapeutic target in cancer treatment, exhibiting potential research interest and clinical benefits. Presently, the novel nanotechnology applied in TME regulation has made huge progress. The nanoparticles (NPs) can be designed as demand to precisely target TME components and to inhibit tumor progression through TME modulation. Moreover, nanotechnology-mediated drug delivery possesses many advantages including prolonged circulation time, enhanced bioavailability and decreased toxicity over traditional therapeutic modality. In this review, update information on TME remodeling through NPs-based targeted drug delivery strategies for anticancer therapy is summarized.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yijun Mei
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yan Shen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Qiaqia Xiao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Yonggang Xu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jie Shao
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.,NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Zihao Cai
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| |
Collapse
|
21
|
Sun J, Ogunnaike EA, Jiang X, Chen Z. Nanotechnology lights up the antitumor potency by combining chemotherapy with siRNA. J Mater Chem B 2021; 9:7302-7317. [PMID: 34382987 DOI: 10.1039/d1tb01379c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanotechnology-based combination anticancer therapy offers novel approaches to overcome the limitations of single-agent administration. The emerging siRNA technology combined with chemotherapy has shown considerable promise in anticancer therapy. There are three main challenges in the fabrication of siRNA/chemotherapeutic drug co-loaded nanovectors: adequate cargo protection, precise targeted delivery, and site-specific cargo release. This review presents a summary of the nanosystems that have recently been developed for co-delivering siRNA and chemotherapeutic drugs. Their combined therapeutic effects are also discussed.
Collapse
Affiliation(s)
- Jian Sun
- College of Nursing, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
| | - Edikan Archibong Ogunnaike
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Xing Jiang
- College of Nursing, Nanjing University of Chinese Medicine, Nanjing, P. R. China.
| | - Zhaowei Chen
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou, P. R. China. and College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P. R. China.
| |
Collapse
|
22
|
Emerging nanomedicine-based therapeutics for hematogenous metastatic cascade inhibition: Interfering with the crosstalk between "seed and soil". Acta Pharm Sin B 2021; 11:2286-2305. [PMID: 34522588 PMCID: PMC8424221 DOI: 10.1016/j.apsb.2020.11.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/18/2020] [Accepted: 09/28/2020] [Indexed: 12/26/2022] Open
Abstract
Despite considerable progresses in cancer treatment, tumor metastasis is still a thorny issue, which leads to majority of cancer-related deaths. In hematogenous metastasis, the concept of “seed and soil” suggests that the crosstalk between cancer cells (seeds) and premetastatic niche (soil) facilitates tumor metastasis. Considerable efforts have been dedicated to inhibit the tumor metastatic cascade, which is a highly complicated process involving various pathways and biological events. Nonetheless, satisfactory therapeutic outcomes are rarely observed, since it is a great challenge to thwart this multi-phase process. Recent advances in nanotechnology-based drug delivery systems have shown great potential in the field of anti-metastasis, especially compared with conventional treatment methods, which are limited by serious side effects and poor efficacy. In this review, we summarized various factors involved in each phase of the metastatic cascade ranging from the metastasis initiation to colonization. Then we reviewed current approaches of targeting these factors to stifle the metastatic cascade, including modulating primary tumor microenvironment, targeting circulating tumor cells, regulating premetastatic niche and eliminating established metastasis. Additionally, we highlighted the multi-phase targeted drug delivery systems, which hold a better chance to inhibit metastasis. Besides, we demonstrated the limitation and future perspectives of nanomedicine-based anti-metastasis strategies.
Collapse
|
23
|
Glucose oxidase loaded Cu 2+ based metal-organic framework for glutathione depletion/reactive oxygen species elevation enhanced chemotherapy. Biomed Pharmacother 2021; 141:111606. [PMID: 34153849 DOI: 10.1016/j.biopha.2021.111606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/29/2021] [Accepted: 04/12/2021] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION The development of multidrug resistance (MDR) is a major cause for the failure of chemotherapy, which requires the aid of nanomedicine. METHODS Here in our study, a Cu2+ based metal-organic framework (COF) was firstly developed and employed as a carrier for the delivery of glucose oxidase (GOx) and doxorubicin (Dox) (COF/GOx/Dox) for the therapy of MDR lung cancers. RESULTS Our results showed that the GOx can catalyze glucose and produce H2O2. In the mean time, the Cu2+ can react with GSH and then transform into Cu+, which resulted in GSH depletion. Afterwards, the produced Cu+ and H2O2 trigger Fenton reaction to generate ROS to damage the redox equilibrium of cancer cells. Both effects contributed to the reverse of MDR in A549/Dox cells and finally resulted in significantly enhanced in vitro/in vivo anticancer performance. DISCUSSION The combination of glutathione depletion/reactive oxygen species elevation might be a promising strategy to enhance the efficacy of chemotherapy and reverse MDR in cancers.
Collapse
|
24
|
Alsaigh T, Di Bartolo BA, Mulangala J, Figtree GA, Leeper NJ. Bench-to-Bedside in Vascular Medicine: Optimizing the Translational Pipeline for Patients With Peripheral Artery Disease. Circ Res 2021; 128:1927-1943. [PMID: 34110900 PMCID: PMC8208504 DOI: 10.1161/circresaha.121.318265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peripheral arterial disease is a growing worldwide problem with a wide spectrum of clinical severity and is projected to consume >$21 billion per year in the United States alone. While vascular researchers have brought several therapies to the clinic in recent years, few of these approaches have leveraged advances in high-throughput discovery screens, novel translational models, or innovative trial designs. In the following review, we discuss recent advances in unbiased genomics and broader omics technology platforms, along with preclinical vascular models designed to enhance our understanding of disease pathobiology and prioritize targets for additional investigation. Furthermore, we summarize novel approaches to clinical studies in subjects with claudication and ischemic ulceration, with an emphasis on streamlining and accelerating bench-to-bedside translation. By providing a framework designed to enhance each aspect of future clinical development programs, we hope to enrich the pipeline of therapies that may prevent loss of life and limb for those with peripheral arterial disease.
Collapse
Affiliation(s)
- Tom Alsaigh
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Belinda A. Di Bartolo
- Cardiothoracic and Vascular Health, Kolling Institute and Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia
| | | | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute and Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
25
|
Chen X, Jin R, Jiang Q, Bi Q, He T, Song X, Barz M, Ai H, Shuai X, Nie Y. Delivery of siHIF-1α to Reconstruct Tumor Normoxic Microenvironment for Effective Chemotherapeutic and Photodynamic Anticancer Treatments. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100609. [PMID: 34032365 DOI: 10.1002/smll.202100609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/17/2021] [Indexed: 06/12/2023]
Abstract
The tumor hypoxic microenvironment not only induces genetic and epigenetic changes in tumor cells, immature vessels formation for oxygen demand, but also compromises the efficiency of therapeutic interventions. On the other hand, conventional therapeutic approaches which kill tumor cells or destroy tumor blood vessels to block nutrition and oxygen supply usually facilitate even harsher microenvironment. Thus, simultaneously relieving the strained response of tumor cells and blood vessels represents a promising strategy to reverse the adverse tumor hypoxic microenvironment. In the present study, an integrated amphiphilic system (RSCD) is designed based on Angiotensin II receptor blocker candesartan for siRNA delivery against the hypoxia-inducible factor-1 alpha (HIF-1α), aiming at both vascular and cellular "relaxation" to reconstruct a tumor normoxic microenvironment. Both in vitro and in vivo studies have confirmed that the hypoxia-inducible HIF-1α expression is down-regulated by 70% and vascular growth is inhibited by 60%. The "relaxation" therapy enables neovascularization with more complete and organized structures to obviously increase the oxygen level inside tumor, which results in a 50% growth inhibition. Moreover, reconstruction of tumor microenvironment enhances tumor-targeted drug delivery, and significantly improves the chemotherapeutic and photodynamic anticancer treatments.
Collapse
Affiliation(s)
- Xiaobing Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qian Jiang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Qunjie Bi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Ting He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xu Song
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Matthias Barz
- Leiden Academic Center for Drug Research (LACDR), Leiden University, Einsteinweg 55, Leiden, 2333 CC, The Netherlands
- Institute of Organic Chemistry, Johannes Gutenberg-University, Mainz Duesbergweg 10-14, 55099, Mainz, Germany
| | - Hua Ai
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xintao Shuai
- PCFM Lab of Ministry of Education, School of Chemistry and Chemical Engineering, Sun Yat-sen University, Guangzhou, 510275, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
26
|
Luo XM, Yan C, Feng YM. Nanomedicine for the treatment of diabetes-associated cardiovascular diseases and fibrosis. Adv Drug Deliv Rev 2021; 172:234-248. [PMID: 33417981 DOI: 10.1016/j.addr.2021.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/25/2020] [Accepted: 01/01/2021] [Indexed: 02/08/2023]
Abstract
Cardiomyopathy and fibrosis are the main causes of heart failure in diabetes patients. For therapeutic purposes, a delivery system is required to enhance antidiabetic drug efficacy and specifically target profibrotic pathways in cardiomyocytes. Nanoparticles (NPs) have distinct advantages, including biocompatibility, bioavailability, targeting efficiency, and minimal toxicity, which make them ideal for antidiabetic treatment. In this review, we overview the latest information on the pathogenesis of cardiomyopathy and fibrosis in diabetes patients. We summarize how NP applications improve insulin and liraglutide efficacy and their sustained release upon oral administration. We provide a comprehensive review of the results of NP clinical trials in diabetes patients and of animal studies investigating the effects of NP-mediated anti-fibrotic treatments. Collectively, the application of advanced NP delivery systems in the treatment of cardiomyopathy and fibrosis in diabetes patients is a promising and innovative therapeutic strategy.
Collapse
|
27
|
Li Q, Zhou R, Sun Y, Xiao D, Liu M, Zhao D, Peng S, Chen Y, Lin Y. Synthesis and Antitumor Application of Antiangiogenetic Gold Nanoclusters. ACS APPLIED MATERIALS & INTERFACES 2021; 13:11708-11720. [PMID: 33656845 DOI: 10.1021/acsami.1c01164] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Conventional antiangiogenetic inhibitors suffered from poor delivery problems that result in unsatisfactory antitumor treatment efficacy. Although the liposomes or nanomaterial-based delivery systems can improve the therapeutic efficacy of antiangiogenic molecules, the assembly process is far too complex. Herein, a nanomaterial or a new nanodrug that could work without the help of a carrier and could be easily synthesized is needed. Au nanoclusters (AuNCs) are a kind of ideal nanostructures that could spontaneously enter into the cell and could be synthesized by a relatively easy one-pot method. Here, changing the traditional ligand glutathione (GSH) into an anti-Flt1 peptide (AF) has enriched the newly synthesized AF@AuNCs with targeted antiangiogenic properties. Based on the specific binding between AF and vascular endothelial growth factor receptor 1 (VEGFR1), the interaction between VEGFR1 and its ligands could be blocked. Furthermore, the expression of VEGFR2 could be downregulated. Compared with pure AF peptide- and GSH-participated AuNCs (GSH@AuNCs), AF@AuNCs were more effective in inhibiting both tube formation and migration of the endothelial cells in vitro. Furthermore, the in vivo chick embryo chorioallantoic membrane (CAM) experiment and antitumor experiment were conducted to further verify the enhanced antiangiogenesis and tumor inhibition effect of AF@AuNCs. Our findings provide promising evidence of a carrier-free nanodrug for tumors and other vascular hyperproliferative diseases.
Collapse
Affiliation(s)
- Qirong Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- West China School of Stomatology, Oral Pathology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yue Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mengting Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Dan Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuanglin Peng
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Southwest Medical University, Luzhou 646000, China
| | - Yu Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- West China School of Stomatology, Oral Pathology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- College of Biomedical Engineering, Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Yang Y, Han Y, Sun Q, Cheng J, Yue C, Liu Y, Song J, Jin W, Ding X, de la Fuente JM, Ni J, Wang X, Cui D. Au-siRNA@ aptamer nanocages as a high-efficiency drug and gene delivery system for targeted lung cancer therapy. J Nanobiotechnology 2021; 19:54. [PMID: 33627152 PMCID: PMC7905599 DOI: 10.1186/s12951-020-00759-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Gene and chemical therapy has become one of the rising stars in the field of molecular medicine during the last two decades. However, there are still numerous challenges in the development of efficient, targeted, and safe delivery systems that can avoid siRNA degradation and reduce the toxicity and adverse effects of chemotherapy medicine. RESULTS In this paper, a highly efficient AS1411 aptamer modified, dsDNA and MMP-2 cleavable peptide-fabricated gold nanocage vehicle, which could load doxorubicin hydrochloride (DOX) and siRNAs to achieve a combination of tumor responsive genetic therapy, chemotherapy, and photothermal treatment is presented. Our results show that this combined treatment achieved targeted gene silencing and tumor inhibition. After nearly one month of treatment with DOX-loaded Au-siRNA-PAA-AS1411 nanoparticles with one dose every three days in mice, a synergistic effect promoting the eradication of long-lived tumors was observed along with an increased survival rate of mice. The combined genetic, chemotherapeutic, and photothermal treatment group exhibited more than 90% tumor inhibition ratio (tumor signal) and a ~ 67% survival rate compared with a 30% tumor inhibition ratio and a 0% survival rate in the passive genetic treatment group. CONCLUSIONS The development of nanocarriers with double-stranded DNA and MMP-2 cleavable peptides provides a new strategy for the combined delivery of gene and chemotherapy medicine. Au-siRNA-PAA-AS1411 exerts high anticancer activities on lung cancer, indicating immense potentials for clinical application.
Collapse
Affiliation(s)
- Yuming Yang
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240, People's Republic of China
| | - Yu Han
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Qiuyang Sun
- Pediatric Neurological Disease Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Number 1665, Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Jin Cheng
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Caixia Yue
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200240, Shanghai, People's Republic of China
| | - Yanlei Liu
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Jie Song
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240, People's Republic of China
| | - Weilin Jin
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240, People's Republic of China
| | - Xianting Ding
- School of Biomedical Engineering, Shanghai Jiao Tong University, 200240, Shanghai, People's Republic of China
| | - Jesús M de la Fuente
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
- Instituto de Nanociencia de Aragon (INA), Universidad de Zaragoza, Zaragoza, 50018, Spain
| | - Jian Ni
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China
| | - Xiaoqiang Wang
- Pediatric Neurological Disease Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Number 1665, Kongjiang Road, Shanghai, 200092, People's Republic of China.
| | - Daxiang Cui
- Institute of Nano Biomedicine and Engineering, Key Laboratory for Thin Film and Microfabrication Technology of the Ministry of Education, Shanghai Engineering Research Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science & Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, People's Republic of China.
- National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan RD, Shanghai, 200240, People's Republic of China.
| |
Collapse
|
29
|
Xiao J, Cong H, Wang S, Yu B, Shen Y. Recent research progress in the construction of active free radical nanoreactors and their applications in photodynamic therapy. Biomater Sci 2021; 9:2384-2412. [PMID: 33576752 DOI: 10.1039/d0bm02013c] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Photodynamic therapy is the most important treatment strategy in free radical therapy. However, tumor microenvironment hypoxia is a key obstacle in PDT. In order to overcome this obstacle, the strategy of in situ production of O2/radicals by catalytic reaction in solid tumors was proposed. In recent years, it has been found that there are many oxygen-independent carbon-based free radicals that can generate toxic active free radicals under laser irradiation and lead to tumor cell death. Based on the rational design of multifunctional nano-medicine, the active free radical nano-generator has opened up a new way for the highly developed nanotechnology and tumor cooperative therapy to improve the therapeutic effect. In this paper, the research status of active free radical nano-generators, especially reactive oxygen species, including the construction mechanism of active free radical nanomaterials, is reviewed and the application of free radical nano-generators in tumor therapy is emphasized.
Collapse
Affiliation(s)
- Jingyuan Xiao
- Institute of Biomedical Materials and Engineering, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China.
| | | | | | | | | |
Collapse
|
30
|
Singh A, Hua Hsu M, Gupta N, Khanra P, Kumar P, Prakash Verma V, Kapoor M. Derivatized Carbon Nanotubes for Gene Therapy in Mammalian and Plant Cells. Chempluschem 2021; 85:466-475. [PMID: 32159284 DOI: 10.1002/cplu.201900678] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/17/2020] [Indexed: 01/06/2023]
Abstract
The concept of gene vectors for therapeutic applications has been known for several years, but it is far from revealing its actual potential. With the advent of hollow cylindrical carbon nanomaterials such as carbon nanotubes (CNTs), researchers have invented several new tools to deliver genes at the required site of action in mammalian and plant cells. The ease of diversified functionalization has allowed CNTs to be by far the most adaptable non-viral vector for gene therapy. This Minireview addresses the dexterity with which CNTs undergo surface modifications and their applications as a potent vector in gene therapy of humans and plants. Specifically, we will discuss the new tools that scientific communities have invented to achieve gene therapy using plasmid DNA, RNA silencing, suicide gene therapy, and plant genetic engineering. Additionally, we will shed some light on the mechanism of gene transportation using carbon nanotubes in cancer cells and plants.
Collapse
Affiliation(s)
- Adhish Singh
- Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, 140401, India
| | - Ming Hua Hsu
- National Changhua University of Education, Changhua, 500, R.O.C. Taiwan
| | - Neeraj Gupta
- Department of Chemistry, Shoolni University, Solon, H.P., 173229, India
| | - Partha Khanra
- Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, 140401, India
| | - Pankaj Kumar
- Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, 140401, India
| | - Ved Prakash Verma
- Department of Chemistry, Banasthali University, Newai-Jodhpuriya Road, Vanasthali, 304022, India
| | - Mohit Kapoor
- Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, 140401, India
| |
Collapse
|
31
|
Li XY, Shi LX, Yao XM, Jing M, Li QQ, Wang YL, Li QS. Functional vinorelbine plus schisandrin B liposomes destroying tumor metastasis in treatment of gastric cancer. Drug Dev Ind Pharm 2021; 47:100-112. [PMID: 33295825 DOI: 10.1080/03639045.2020.1862169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Gastric cancer is one of the leading causes of cancer-related death worldwide with a poor prognosis. Gastric cancer is usually treated with surgery and chemotherapy, accompanied by a high rate of metastasis and recurrence. In this paper, R8 (RRRRRRRR) modified vinorelbine plus schisandrin B liposomes had been successfully constructed for treating gastric cancer. In the liposomes, R8 was used to enhance the intracellular uptake, schisandrin B was incorporated into liposomes for inhibiting tumor cells metastasis, and vinorelbine was encapsulated into liposomes as antitumor drugs. Studies were performed on BGC-823 cells in vitro and were verified in the BGC-823 cell xenografts nude mice in vivo. Results in vitro demonstrated that the targeting liposomes could induce BGC-823 cells apoptosis, inhibit the metastasis of tumor cells, and increase targeting effects to tumor cells. Meanwhile, action mechanism studies showed that the targeting liposomes could down-regulate VEGF, VE-Cad, HIF-1a, PI3K, MMP-2, and FAK to inhibit tumor metastasis. In vivo results exhibited that the targeting liposomes displayed an obvious antitumor efficacy by accumulating selectively in tumor site and induce tumor cell apoptosis. Hence, R8 modified vinorelbine plus schisandrin B liposomes might provide a safe and efficient therapy strategy for gastric cancer.
Collapse
Affiliation(s)
- Xiu-Ying Li
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Luan-Xia Shi
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Xue-Min Yao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ming Jing
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Qin-Qing Li
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Ying-Li Wang
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qing-Shan Li
- School of Pharmacy, Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
32
|
Zhang M, Chen X, Radacsi N. New tricks of old drugs: Repurposing non-chemo drugs and dietary phytochemicals as adjuvants in anti-tumor therapies. J Control Release 2020; 329:96-120. [PMID: 33259852 DOI: 10.1016/j.jconrel.2020.11.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
Combination therapy has long been applied to enhance therapeutic effect and deal with the occurrence of multi-drug resistance in cancer treatment. However, the overlapping toxicity of multiple anticancer drugs to healthy tissues and increasing financial burden on patients emerged as major concerns. As promising alternatives to chemo agents, repurposed non-chemo drugs and dietary phytochemicals have been investigated as adjuvants to conventional anti-tumor therapeutics, offering a safe and economic strategy for combination therapy. In this review, we aim to highlight the advances in research about combination therapy using conventional therapeutics and repurposed drugs or phytochemicals for an enhanced anti-tumor efficacy, along with the mechanisms involved in the synergism. Beyond these, we outlined the potential challenges and solutions for clinical translation of the proposed combination therapy, providing a safe and affordable strategy to improve the reach of cancer therapy to low income regions with such new tricks of old drugs.
Collapse
Affiliation(s)
- Mei Zhang
- School of Engineering, Institute for Materials and Processes, University of Edinburgh, Robert Stevenson Road, Edinburgh EH9 3FB, United Kingdom; School of Engineering, Institute for Bioengineering, University of Edinburgh, The King's Buildings, Edinburgh EH9 3JL, United Kingdom.
| | - Xianfeng Chen
- School of Engineering, Institute for Bioengineering, University of Edinburgh, The King's Buildings, Edinburgh EH9 3JL, United Kingdom.
| | - Norbert Radacsi
- School of Engineering, Institute for Materials and Processes, University of Edinburgh, Robert Stevenson Road, Edinburgh EH9 3FB, United Kingdom.
| |
Collapse
|
33
|
Li Y, Yu H, Zhao L, Zhu Y, Bai R, Jin Z, Fu Z, Zhang X, Su J, Liu H, Shi X, Han D, Chen Y. Effects of carbon nanotube-mediated Caspase3 gene silencing on cardiomyocyte apoptosis and cardiac function during early acute myocardial infarction. NANOSCALE 2020; 12:21599-21604. [PMID: 33103172 DOI: 10.1039/d0nr05032f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
RNA interference (RNAi) technology can achieve efficient and specific silencing of Caspase3 gene expression, thus providing new options for anti-apoptosis treatment. However, delivering siRNA to specific cells and tissues in the body is a significant challenge. Therefore, we aim to construct a functionalized single-walled carbon nanotube (F-CNT) bound to siRNA from Caspase3. The obtained gene transfer carrier F-CNT-siCas3 not only demonstrated a good water solubility and biocompatibility, but also had a high transfection efficiency of up to 82%, which significantly downregulated the expression level of the Caspase3 gene miRNA and protein in primary cardiomyocytes. Furthermore, it was verified by in vivo experiments that Caspase3 gene silencing had obvious protective effects on myocardial cell apoptosis, ventricular remodeling, and cardiac function in Sprague-Dawley (SD) rats after coronary artery ligation. This study may provide an important theoretical basis for the application of F-CNT in vivo siRNA gene therapy to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Yi Li
- Department of Cardiology, The Third Medical Center, Chinese PLA General Hospital, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Manipulation of immune‒vascular crosstalk: new strategies towards cancer treatment. Acta Pharm Sin B 2020; 10:2018-2036. [PMID: 33304777 PMCID: PMC7714955 DOI: 10.1016/j.apsb.2020.09.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/25/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022] Open
Abstract
Tumor vasculature is characterized by aberrant structure and function, resulting in immune suppressive profiles of tumor microenvironment through limiting immune cell infiltration into tumors, endogenous immune surveillance and immune cell function. Vascular normalization as a novel therapeutic strategy tends to prune some of the immature blood vessels and fortify the structure and function of the remaining vessels, thus improving immune stimulation and the efficacy of immunotherapy. Interestingly, the presence of "immune‒vascular crosstalk" enables the formation of a positive feedback loop between vascular normalization and immune reprogramming, providing the possibility to develop new cancer therapeutic strategies. The applications of nanomedicine in vascular-targeting therapy in cancer have gained increasing attention due to its specific physical and chemical properties. Here, we reviewed the recent advances of effective routes, especially nanomedicine, for normalizing tumor vasculature. We also summarized the development of enhancing nanoparticle-based anticancer drug delivery via the employment of transcytosis and mimicking immune cell extravasation. This review explores the potential to optimize nanomedicine-based therapeutic strategies as an alternative option for cancer treatment.
Collapse
|
35
|
Bhattacharjee S, Brayden DJ. Addressing the challenges to increase the efficiency of translating nanomedicine formulations to patients. Expert Opin Drug Discov 2020; 16:235-254. [PMID: 33108229 DOI: 10.1080/17460441.2021.1826434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nanotechnology is in a growth phase for drug delivery and medical imaging. Nanomaterials with unique properties present opportunities for encapsulation of therapeutics and imaging agents, along with conjugation to ligands for targeting. Favorable chemistry of nanomaterials can create formulations that address critical challenges for therapeutics, such as insolubility and a low capacity to cross the blood-brain-barrier (BBB) and intestinal wall. AREAS COVERED The authors investigate challenges faced during translation of nanomedicines while suggesting reasons as to why some nanoformulations have under-performed in clinical trials. They assess physiological barriers such as the BBB and gut mucus that nanomedicines must overcome to deliver cargos. They also provide an overview with examples of how nanomedicines can be designed to improve localization and site-specific delivery (e.g., encapsulation, bioconjugation, and triggered-release). EXPERT OPINION There are examples where nanomedicines have demonstrated improved efficacy of payload in humans; however, most of the advantages conferred were in improved pharmacokinetics and reduced toxicity. Problematic data show susceptibility of nanoformulations against natural protective mechanisms present in the body, including distribution impediment by physiological barriers and activation of the reticuloendothelial system. Further initiatives should address current challenges while expanding the scope of nanomedicine into advanced biomedical imaging and antibiotic delivery.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland
| | - David J Brayden
- School of Veterinary Medicine, University College Dublin (UCD), Belfield, Dublin, Ireland.,Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Belfield, Dublin, Ireland
| |
Collapse
|
36
|
Supe S, Upadhya A, Singh K. Role of small interfering RNA (siRNA) in targeting ocular neovascularization: A review. Exp Eye Res 2020; 202:108329. [PMID: 33198953 DOI: 10.1016/j.exer.2020.108329] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/30/2020] [Accepted: 10/21/2020] [Indexed: 12/16/2022]
Abstract
Ocular neovascularization (NV) plays a central role in the pathogenesis of various ocular diseases including diabetic retinopathy, age-related macular degeneration, retinoblastoma, retinitis pigmentosa and may lead to loss of vision if not controlled in time. Several clinical trials elucidate the central role of vascular endothelial growth factor (VEGF) in the pathogenesis of the ocular neovascularization. The advent and extensive use of ocular anti-VEGF therapy heralded a new age in the treatment of retinal vascular and exudative diseases. RNA interference (RNAi) can be used to inhibit the in-vitro and in-vivo expression of specific genes and thus provides an extremely useful method for investigating gene activity with minimal toxicity. siRNA targeting VEGF overcomes many drawbacks associated with the conventional treatment available for the treatment of ocular neovascularization. However, delivery methods that protect the siRNA against degradation and are appropriate for long-term care will help increase the effectiveness of RNAi-based anti-VEGF ocular therapies. Several nanotechnology approaches have been explored by formulation scientists for delivery of siRNA to the eye; targeting particularly VEGF for the treatment of NV. This review mainly focuses on current updates in various pre-clinical and clinical siRNA strategies for targeting VEGF involved in the development of ocular neovascularization.
Collapse
Affiliation(s)
- Shibani Supe
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, 400056, Maharashtra, India
| | - Archana Upadhya
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, 400056, Maharashtra, India
| | - Kavita Singh
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, 400056, Maharashtra, India.
| |
Collapse
|
37
|
Cai XJ, Wang Z, Xu YY, Yang GY, Zhang RY, Wang Y. Candesartan treatment enhances liposome penetration and anti-tumor effect via depletion of tumor stroma and normalization of tumor vessel. Drug Deliv Transl Res 2020; 11:1186-1197. [PMID: 32822012 DOI: 10.1007/s13346-020-00842-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The poor penetration of nanoparticles in solid tumors has been a critical factor limiting the clinical benefits of nanomedicine. Therefore, we depleted the dense extracellular matrix (ECM) and normalized tumor vessels to enhance drug delivery and therapeutic efficacy. We used candesartan as an angiotensin system inhibitor, which reduced ECM content and facilitated "vascular normalization" by targeting the angiotensin-signaling axis, resulting in improved anti-cancer therapeutic effects. We also combined candesartan with PEGylated liposome-encapsulated zoledronic acid (ZOL) (PEG-ZOL-LPs) to assess how this affected anti-tumor therapy. Our findings indicated that the migration of 4T1 mouse breast cancer cells was inhibited by candesartan. Moreover, the ECM depletion (including collagen I and hyaluronan) by candesartan was achieved through the downregulation of TGF-β1 in vitro, consistent with in vivo results. Furthermore, treatment groups that received candesartan also had significantly decreased tumor vessel permeability and proportions of circulating endothelial progenitor cells (CEPCs) in the serum, which resulted in normalization of tumor vasculature and improved delivery of PEG-ZOL-LPs. Finally, the positive effect candesartan in terms of tumor growth was found not to have an impact of the efficacy of the PEG-ZOL-LPs treatment. This unexpected lack of effect of candesartan on the performance of PEG-ZOL-LPs would be due to dynamics of the effect of both treatments. It might be possible that a different protocol of administration could lead to a synergistic effect. Graphical abstract The schematic illustration showed that candesartan favored depletion of tumor stroma and tumor vascular normalization to improve the anti-cancer efficacy of PEG-ZOL-LPs.
Collapse
Affiliation(s)
- Xin-Jun Cai
- Department of Pharmacy, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang, People's Republic of China.
| | - Zeng Wang
- Department of Pharmacy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Ying-Ying Xu
- Department of Pharmacy, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Gao-Yi Yang
- Department of Ultrasoud, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Ruo-Ying Zhang
- Department of Pharmacy, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang, People's Republic of China
| | - Yu Wang
- Department of Pharmacy, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou Red Cross Hospital, Hangzhou, 310003, Zhejiang, People's Republic of China
| |
Collapse
|
38
|
Kargozar S, Baino F, Hamzehlou S, Hamblin MR, Mozafari M. Nanotechnology for angiogenesis: opportunities and challenges. Chem Soc Rev 2020; 49:5008-5057. [PMID: 32538379 PMCID: PMC7418030 DOI: 10.1039/c8cs01021h] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis plays a critical role within the human body, from the early stages of life (i.e., embryonic development) to life-threatening diseases (e.g., cancer, heart attack, stroke, wound healing). Many pharmaceutical companies have expended huge efforts on both stimulation and inhibition of angiogenesis. During the last decade, the nanotechnology revolution has made a great impact in medicine, and regulatory approvals are starting to be achieved for nanomedicines to treat a wide range of diseases. Angiogenesis therapies involve the inhibition of angiogenesis in oncology and ophthalmology, and stimulation of angiogenesis in wound healing and tissue engineering. This review aims to summarize nanotechnology-based strategies that have been explored in the broad area of angiogenesis. Lipid-based, carbon-based and polymeric nanoparticles, and a wide range of inorganic and metallic nanoparticles are covered in detail. Theranostic and imaging approaches can be facilitated by nanoparticles. Many preparations have been reported to have a bimodal effect where they stimulate angiogenesis at low dose and inhibit it at higher doses.
Collapse
Affiliation(s)
- Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, 917794-8564 Mashhad, Iran
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 101 29 Torino, Italy
| | - Sepideh Hamzehlou
- Hematology/Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
| | - Masoud Mozafari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Carbon nanotube embedded cyclodextrin polymer derived injectable nanocarrier: A multiple faceted platform for stimulation of multi-drug resistance reversal. Carbohydr Polym 2020; 247:116751. [PMID: 32829867 DOI: 10.1016/j.carbpol.2020.116751] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 11/21/2022]
Abstract
A combination of cocktail chemotherapy (CCT), photothermal therapy (PTT) and inhibition of angiogenesis was investigated as an effective approach to combat major challenges of multidrug resistance and non-targeted drug delivery encountered in conventional cancer therapy. An injectable nanocarrier was developed through functionalization of carbon nanotubes (CNTs) with rationally modified carbohydrate (β-Cyclodextrin-CD) derived pH and thermo responsive polymer. Embedding CNT to CD polymer offers a nanocarrier which effectively demonstrated CCT, high NIR triggered photothermal efficiency, anti-angiogenic potential for selective tumor homing as well as enhanced multi-drug resistance (MDR) reversal with minimal toxic effects on normal cells. The simultaneously loading with curcumin and doxorubicin hydrochloride exhibited synergistic effect for triggering antitumor effect in vitro and demonstrated down regulation of growth factors associated with angiogenesis ex-ovo. In-vivo studies ascertained that the nanocarrier synthesized with the rational for MDR reversal can lead to enhanced cancer cell death via multiple approaches.
Collapse
|
40
|
Far-reaching advances in the role of carbon nanotubes in cancer therapy. Life Sci 2020; 257:118059. [PMID: 32659368 DOI: 10.1016/j.lfs.2020.118059] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022]
Abstract
Cancer includes a group of diseases involving unregulated cell growth with the potential to invade or expand to other parts of the body, resulting in an estimate of 9.6 million deaths worldwide in 2018. Manifold studies have been conducted to design more efficacious techniques for cancer therapy due to the inadequacy of conventional treatments including chemotherapy, surgery, and radiation therapy. With the advances in the biomedical applications of nanotechnology-based systems, nanomaterials have gained increasing attention as promising vehicles for targeted cancer therapy and optimizing treatment outcomes. Owing to their outstanding thermal, electrical, optical and chemical properties, carbon nanotubes (CNTs) have been profoundly studied to explore the various perspectives of their application in cancer treatment. The current study aims to review the role of CNTs whether as a carrier or mediator in cancer treatment for enhancing the efficacy as well as the specificity of therapy and reducing adverse side effects. This comprehensive review indicates that CNTs have the capability to be the next generation nanomaterials to actualize noninvasive targeted eradication of tumors. However, further studies are needed to evaluate the consequences of their biomedical application before the transition into clinical trials, since possible adverse effects of CNTs on biological systems have not been clearly understood.
Collapse
|
41
|
Navarro-Partida J, Altamirano-Vallejo JC, Lopez-Naranjo EJ, Gonzalez-De la Rosa A, Manzano-Ramírez A, Apatiga-Castro LM, Armendáriz-Borunda J, Santos A. Topical Triamcinolone Acetonide-Loaded Liposomes as Primary Therapy for Macular Edema Secondary to Branch Retinal Vein Occlusion: A Pilot Study. J Ocul Pharmacol Ther 2020; 36:393-403. [PMID: 32564664 DOI: 10.1089/jop.2019.0143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Purpose: To explore safety and therapeutic efficacy of a topical ophthalmic triamcinolone acetonide-loaded liposome formulation (TA-LF) as primary therapy in patients with macular edema (ME) secondary to branch retinal vein occlusion (BRVO). Methods: Twelve eyes of 12 patients with ME secondary to BRVO were exposed to a topical instillation of 1 drop of TA-LF (TA 0.2%) 6 times a day for 12 weeks to evaluate safety and efficacy. Best corrected visual acuity (BCVA) intraocular pressure (IOP), slit lamp examination, and central foveal thickness (CFT) were analyzed at every visit. In addition, the morphology of TA-LF was analyzed using scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Results: Patients presented a significant improvement of BCVA and CFT without significant IOP modification (P = 0.94). Treated eyes showed BCVA improvement from 40 ± 12.05 to 64.83 ± 15.97 letters and CFT reduction from 682.91 ± 278.60 to 271.58 ± 57.66 μm after 12 weeks of TA-LF therapy (P < 0.001). No adverse events, including IOP rising, were registered. SEM analysis of liposomal formulations showed that liposome (LP) size depends on its concentration. As the concentration of TA increased, the average size of LPs and the number of larger particles increased as well. TEM study displayed that LP formulation efficiently solubilizes TA crystals in nanoparticles and encapsulates them. Conclusion: LPs can function as nanocarriers of TA and they could be used as topical ophthalmic primary therapy instead of intravitreal drugs in patients with ME secondary to BRVO.
Collapse
Affiliation(s)
- Jose Navarro-Partida
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan, Mexico.,Centro de Retina Medica y Quirúrgica, S.C., Centro Medico Puerta de Hierro, Zapopan, Mexico
| | - Juan Carlos Altamirano-Vallejo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan, Mexico.,Centro de Retina Medica y Quirúrgica, S.C., Centro Medico Puerta de Hierro, Zapopan, Mexico
| | | | - Alejandro Gonzalez-De la Rosa
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan, Mexico.,Centro de Retina Medica y Quirúrgica, S.C., Centro Medico Puerta de Hierro, Zapopan, Mexico
| | | | - Luis Miguel Apatiga-Castro
- Universidad Nacional Autonoma de Mexico (UNAM), Centro de Física Aplicada y Tecnología Avanzada, Querétaro, Mexico
| | - Juan Armendáriz-Borunda
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan, Mexico.,Instituto de Biología Molecular y Terapia Génica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Mexico
| | - Arturo Santos
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan, Mexico.,Centro de Retina Medica y Quirúrgica, S.C., Centro Medico Puerta de Hierro, Zapopan, Mexico
| |
Collapse
|
42
|
Cao Y, Luo Y. Pharmacological and toxicological aspects of carbon nanotubes (CNTs) to vascular system: A review. Toxicol Appl Pharmacol 2019; 385:114801. [PMID: 31678607 DOI: 10.1016/j.taap.2019.114801] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/15/2019] [Accepted: 10/30/2019] [Indexed: 01/12/2023]
Abstract
Carbon nanotubes (CNTs) are novel carbon based nanomaterials (NMs) that could be used in many areas ranging from electronics to biotechnology. The present review summarized pharmacological and toxicological aspects of CNTs to vascular systems, because the vascular systems are important targets for CNTs during manufacturing process, daily contact and biomedical uses. Functionalized CNTs could be used as novel nanoplateforms to regulate angiogenesis for cancer therapy, as well as nanocarriers to cross blood brain barrier (BBB), one of the major obstacles to prevent the entering of therapeutic substances into brains. However, it has also been shown that inhalational or intravenous contact with CNTs might induce adverse vascular effects, such as progression of atherosclerotic plaque, vasomotor dysfunction, and changes of blood pressure and/or heart rate in laboratory animals, although currently there are only limited reports obtained from CNT-exposed human beings and the results are inconclusive. The mechanisms associated with the vascular toxicity of CNTs remain poorly understood, and it appears that multiple signaling pathways are likely to be involved. The toxicity of CNTs to vascular systems might be reduced by controlling the physicochemical properties of CNTs, particularly lengths, diameters and surface chemistry. At present, the beneficial and adverse effects of CNTs to vascular systems are still largely unknown and require further extensive studies.
Collapse
Affiliation(s)
- Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Lab of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China.
| | - Yingmei Luo
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Lab of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan 411105, PR China
| |
Collapse
|
43
|
Li F, Wang Y, Chen WL, Wang DD, Zhou YJ, You BG, Liu Y, Qu CX, Yang SD, Chen MT, Zhang XN. Co-delivery of VEGF siRNA and Etoposide for Enhanced Anti-angiogenesis and Anti-proliferation Effect via Multi-functional Nanoparticles for Orthotopic Non-Small Cell Lung Cancer Treatment. Am J Cancer Res 2019; 9:5886-5898. [PMID: 31534526 PMCID: PMC6735374 DOI: 10.7150/thno.32416] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Targeting tumor angiogenesis pathway via VEGF siRNA (siVEGF) has shown great potential in treating highly malignant and metastatic non-small cell lung cancer (NSCLC). However, anti-angiogenic monotherapy lacked sufficient antitumor efficacy which suffered from malignant tumor proliferation. Therefore, the combined application of siVEGF and chemotherapeutic agents for simultaneous targeting of tumor proliferation and angiogenesis has been a research hotspot to explore a promising NSCLC therapy regimen. Methods: We designed, for the first time, a rational therapy strategy via intelligently co-delivering siVEGF and chemotherapeutics etoposide (ETO) by multi-functional nanoparticles (NPs) directed against the orthotopic NSCLC. These NPs consisted of cationic liposomes loaded with siVEGF and ETO and then coated with versatile polymer PEGylated histidine-grafted chitosan-lipoic acid (PHCL). We then comprehensively evaluated the anti-angiogenic and anti-proliferation efficiency in the in vitro tumor cell model and in bioluminescent orthotopic lung tumor bearing mice model. Results: The NPs co-delivering siVEGF and ETO exhibited tailor-made surface charge reversal features in mimicking tumor extracellular environment with improved internal tumor penetration capacity and higher cellular internalization. Furthermore, these NPs with flexible particles size triggered by intracellular acidic environment and redox environment showed pinpointed and sharp intracellular cargo release guaranteeing adequate active drug concentration in tumor cells. Enhanced VEGF gene expression silencing efficacy and improved tumor cell anti-proliferation effect were demonstrated in vitro. In addition, the PHCL layer improved the stability of these NPs in neutral environment allowing enhanced orthotopic lung tumor targeting efficiency in vivo. The combined therapy by siVEGF and ETO co-delivered NPs for orthotopic NSCLC simultaneously inhibited tumor proliferation and tumor angiogenesis resulting in more significant suppression of tumor growth and metastasis than monotherapy. Conclusion: Combined application of siVEGF and ETO by the multi-functional NPs with excellent and on-demand properties exhibited the desired antitumor effect on the orthotopic lung tumor. Our work has significant potential in promoting combined anti-angiogenesis therapy and chemotherapy regimen for clinical NSCLC treatment.
Collapse
|
44
|
Liu Y, Yu Q, Chang J, Wu C. Nanobiomaterials: from 0D to 3D for tumor therapy and tissue regeneration. NANOSCALE 2019; 11:13678-13708. [PMID: 31292580 DOI: 10.1039/c9nr02955a] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Nanobiomaterials have attracted tremendous attention in the biomedical field. Especially in the past few years, a large number of low dimensional nanobiomaterials, including 0D nanostructures, 1D nanotubes and 2D nanosheets, were employed for tumor therapy due to their optically triggered tumor therapy effects and drug loading capacities. However, these low dimensional nanobiomaterials cannot support cell adhesion and possess poor tissue regeneration ability, thus they are not suitable for application in regenerative medicine. Three dimensional (3D) nanofiber scaffolds have attracted extensive attention in tissue regeneration, including bone, skin, nerve and cardiac tissues, due to their similar extracellular matrix structures. Additionally, many 3D scaffolds displayed bone and cartilage regeneration abilities. Therefore, to obtain materials with both tumor therapy and tissue regeneration abilities, it is meaningful and necessary to develop 3D nanobiomaterials with multifunctions. In this review, we systematically review the research progress of nanobiomaterials with varied dimensional structures including 0D, 1D, 2D and 3D, as well as evolutional functions from single tumor therapy to simultaneous tumor therapy and tissue regeneration. This review may pave the way for developing an interdisciplinary research of nanobiomaterials in combination of tumor therapy and regenerative medicine.
Collapse
Affiliation(s)
- Yaqin Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China. and Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Qingqing Yu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China. and Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jiang Chang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China. and Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, China. and Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
45
|
Panwar N, Soehartono AM, Chan KK, Zeng S, Xu G, Qu J, Coquet P, Yong KT, Chen X. Nanocarbons for Biology and Medicine: Sensing, Imaging, and Drug Delivery. Chem Rev 2019; 119:9559-9656. [DOI: 10.1021/acs.chemrev.9b00099] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nishtha Panwar
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Alana Mauluidy Soehartono
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Kok Ken Chan
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Shuwen Zeng
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
- CINTRA CNRS/NTU/THALES, UMI 3288, Research Techno Plaza, 50 Nanyang Drive, Border X Block, Singapore 637553, Singapore
| | - Gaixia Xu
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Junle Qu
- Key Laboratory of Optoelectronics Devices and Systems of Ministry of Education/Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, P. R. China
| | - Philippe Coquet
- CINTRA CNRS/NTU/THALES, UMI 3288, Research Techno Plaza, 50 Nanyang Drive, Border X Block, Singapore 637553, Singapore
- Institut d’Electronique, de Microélectronique et de Nanotechnologie (IEMN), CNRS UMR 8520—Université de Lille, 59650 Villeneuve d’Ascq, France
| | - Ken-Tye Yong
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
46
|
Li T, Shi S, Goel S, Shen X, Xie X, Chen Z, Zhang H, Li S, Qin X, Yang H, Wu C, Liu Y. Recent advancements in mesoporous silica nanoparticles towards therapeutic applications for cancer. Acta Biomater 2019; 89:1-13. [PMID: 30797106 DOI: 10.1016/j.actbio.2019.02.031] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/18/2019] [Accepted: 02/19/2019] [Indexed: 01/25/2023]
Abstract
Recently, drug delivery systems based on nanotechnology have received great attention in cancer therapeutics and diagnostics since they can not only improve the treatment efficacy but also reduce the side effects. Among them, mesoporous silica nanoparticles (MSNs) with large surface area, high pore volume, tunable pore size, abundant surface chemistry, and acceptable biocompatibility exhibit unique advantages and are considered as promising candidates for cancer diagnosis and therapy. In this review, we update the recent progress on MSN-based systems for cancer treatment purposes. We also discuss the drug loading mechanism of MSNs, stimuli-responsive drug release, and surface modification strategies for improving biocompatibility, and targeting functionalities. STATEMENT OF SIGNIFICANCE: The development of MSN-based delivery systems that can be used in both diagnosis and treatment of cancer has attracted tremendous interest in the past decade. MSN-based delivery systems can improve therapeutic efficacy and reduce cytotoxicity to normal tissue. To further improve the in vivo properties of MSNs and potential translation to the clinics, it is critical to design MSNs with appropriate surface engineering and desirable cancer targeting. This review is intended to provide the readers a comprehensive background of the vast literature till date on silica-based drug delivery systems, and to inspire further innovations in silica nanomedicine in the future.
Collapse
Affiliation(s)
- Tingting Li
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Sixiang Shi
- Department of Cancer Systems Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Shreya Goel
- Department of Materials Science & Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Xue Shen
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiaoxue Xie
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Zhongyuan Chen
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hanxi Zhang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Shun Li
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiang Qin
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hong Yang
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Chunhui Wu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yiyao Liu
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Hospital of Chengdu University of Traditional Chinese Medicine, No.39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China.
| |
Collapse
|
47
|
Yin Y, Lee MS, Lee JE, Lim SY, Kim ES, Jeong J, Kim D, Kim J, Lee DS, Jeong JH. Effective systemic siRNA delivery using dual-layer protected long-circulating nanohydrogel containing an inorganic core. Biomater Sci 2019; 7:3297-3306. [DOI: 10.1039/c9bm00369j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PEG-dex-dopa nanohydrogel containing a CaP/siRNA core could achieve extended circulation with reduced RES accumulation, resulting in increased tumor accumulation.
Collapse
|
48
|
Jiang C, Qi Z, Jia H, Huang Y, Wang Y, Zhang W, Wu Z, Yang H, Liu J. ATP-Responsive Low-Molecular-Weight Polyethylenimine-Based Supramolecular Assembly via Host-Guest Interaction for Gene Delivery. Biomacromolecules 2018; 20:478-489. [PMID: 30516950 DOI: 10.1021/acs.biomac.8b01395] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this work, we report on an ATP-responsive low-molecular-weight polyethylenimine (LMW-PEI)-based supramolecular assembly. It formed via host-guest interaction between PEI (MW = 1.8 kDa)-α-cyclodextrin (α-CD) conjugates and PEI1.8k-phenylboronic acid (PBA) conjugates. The host-guest interaction between PEI1.8k-α-CD and PEI1.8k-PBA was confirmed by the 2D-NOESY chromatogram experiment and competition test. The ATP-responsive property of the supramolecular assembly was evaluated by a series of ATP-triggered degradation and siRNA release studies in terms of fluorescence resonance energy transfer, agarose gel electrophoresis assay, and the time course monitoring of the particle size and morphology. Confocal laser scanning microscopy confirmed the intracellular disassembly of the supramolecular polymer and the release of siRNA. The supramolecular assembly showed high buffering capability and was capable of protecting siRNA from RNase degradation. It had high cytocompatibility according to in vitro cytotoxicity and hemolysis assays. LMW-PEI-based supramolecular assembly facilitated cellular entry of siRNA via energy-dependent endocytosis. Moreover, the assembly/SR-A siRNA polyplexes at N/P ratio of 30 was most effective in knocking down SR-A mRNA and inhibiting uptake of modified LDL. Taken together, this work shows that ATP-responsive LMW-PEI-based supramolecular assembly is a promising gene vector and has potential application in treating atherosclerosis.
Collapse
Affiliation(s)
- Cuiping Jiang
- Department of Pharmaceutics , China Pharmaceutical University , Nanjing , Jiangsu 210009 , People's Republic of China
| | - Zitong Qi
- Department of Pharmaceutics , China Pharmaceutical University , Nanjing , Jiangsu 210009 , People's Republic of China
| | - Hengbo Jia
- Department of Pharmaceutics , China Pharmaceutical University , Nanjing , Jiangsu 210009 , People's Republic of China
| | - Yilei Huang
- Department of Pharmaceutics , China Pharmaceutical University , Nanjing , Jiangsu 210009 , People's Republic of China
| | - Yunbo Wang
- Department of Pharmaceutics , China Pharmaceutical University , Nanjing , Jiangsu 210009 , People's Republic of China
| | - Wenli Zhang
- Department of Pharmaceutics , China Pharmaceutical University , Nanjing , Jiangsu 210009 , People's Republic of China
| | - Zimei Wu
- School of Pharmacy , University of Auckland , Private Bag 92019, Auckland , New Zealand
| | - Hu Yang
- Department of Chemical and Life Science Engineering , Virginia Commonwealth University , Richmond , Virginia 23219 , United States.,Department of Pharmaceutics , Virginia Commonwealth University , Richmond , Virginia 23298 , United States.,Massey Cancer Center, Virginia Commonwealth University , Richmond , Virginia 23298 , United States
| | - Jianping Liu
- Department of Pharmaceutics , China Pharmaceutical University , Nanjing , Jiangsu 210009 , People's Republic of China
| |
Collapse
|
49
|
Chun SH, Yuk JS, Um SH. Regulation of cellular gene expression by nanomaterials. NANO CONVERGENCE 2018; 5:34. [PMID: 30499017 PMCID: PMC6265357 DOI: 10.1186/s40580-018-0166-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 11/15/2018] [Indexed: 06/04/2023]
Abstract
Within a cell there are several mechanisms to regulate gene expression during cellular metabolism, growth, and differentiation. If these do not work properly, the cells will die or develop abnormally and, in some cases, even develop into tumors. Thus, a variety of exogenous and endogenous approaches have been developed that act on essential stages of transcription and translation by affecting the regulation of gene expression in an intended manner. To date, some anticancer strategies have focused on targeting abnormally overexpressed genes termed oncogenes, which have lost the ability to tune gene expression. With the rapid advent of nanotechnology, a few synthetic nanomaterials are being used as gene regulation systems. In many cases, these materials have been employed as nanocarriers to deliver key molecules such as silencing RNAs or antisense oligonucleotides into target cells, but some nanomaterials may be able to effectively modulate gene expression due to their characteristic properties, which include tunable physicochemical properties due to their malleable size and shape. This technology has improved the performance of existing approaches for regulating gene expression and led to the development of new types of advanced regulatory systems. In this short review, we will present some nanomaterials currently used in novel gene regulation systems, focusing on their basic features and practical applications. Based on these findings, it is further envisioned that next-generation gene expression regulation systems involving such nanomaterials will be developed.
Collapse
Affiliation(s)
- Sang Hun Chun
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746 South Korea
| | - Ji Soo Yuk
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746 South Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, Gyeonggi-do 440-746 South Korea
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University, Suwon, Gyeonggi-do 440-746 South Korea
| |
Collapse
|
50
|
Li M, Zhang F, Su Y, Zhou J, Wang W. Nanoparticles designed to regulate tumor microenvironment for cancer therapy. Life Sci 2018; 201:37-44. [PMID: 29577880 DOI: 10.1016/j.lfs.2018.03.044] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/12/2018] [Accepted: 03/21/2018] [Indexed: 02/08/2023]
Abstract
Increasing understanding in tumor pathology reveals that tumor microenvironment (TME), which supports tumor progression and poses barriers for available therapies, takes a great responsibility in inefficient treatment and poor prognosis. In recent years, the versatile nanotechnology employed in TME regulation has made great progress. The nanoparticles (NPs) can be tailored as needed to accurately target TME components by distinguishing healthy tissues from malignancy, and to regulate TME to promote tumor regression. Meanwhile, the emerging microRNAs (miRNAs) demonstrate great potentials for TME regulation, but are regrettably restricted by quick degradation. NPs systems enable the successful delivery of miRNA to TME without the limitation, expanding the application of nucleic acid drug. In this review, we summarized recent NPs-based strategies aiming at regulating TME in different ways, including anti-angiogenesis, extracellular matrix (ECM) remodeling, tumor-associated fibroblasts (TAFs) treatment and tumor-associated macrophages (TAMs) treatment, along with the miRNAs-loaded NPs for TME regulation. Catching and utilizing the features of TME for NPs design can contribute to reversing drug-resistance, optimized drug distribution, and eventually more efficient cancer therapy.
Collapse
Affiliation(s)
- Min Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Fangrong Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Yujie Su
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, China.
| |
Collapse
|