1
|
Wang Y, Liao Y, Zhang YJ, Wu XH, Qiao ZY, Wang H. Self-Assembled Peptide with Morphological Structure for Bioapplication. Biomacromolecules 2024; 25:6367-6394. [PMID: 39297513 DOI: 10.1021/acs.biomac.4c01179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Peptide materials, such as self-assembled peptide materials, are very important biomaterials. Driven by multiple interaction forces, peptide molecules can self-assemble into a variety of different macroscopic forms with different properties and functions. In recent years, the research on self-assembled peptides has made great progress from laboratory design to clinical application. This review focuses on the different morphologies, including nanoparticles, nanovesicles, nanotubes, nanofibers, and others, formed by self-assembled peptide. The mechanisms and applications of the morphology transformation are also discussed in this paper, and the future direction of self-assembled nanomaterials is envisioned.
Collapse
Affiliation(s)
- Yu Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Yusi Liao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, P. R. China
| | - Ying-Jin Zhang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Xiu-Hai Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin150081, P. R. China
| | - Zeng-Ying Qiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Hao Wang
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No.11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Guangxi Medical University, Nanning 530021, P. R. China
| |
Collapse
|
2
|
Rajaram J, Mende LK, Kuthati Y. A Review of the Efficacy of Nanomaterial-Based Natural Photosensitizers to Overcome Multidrug Resistance in Cancer. Pharmaceutics 2024; 16:1120. [PMID: 39339158 PMCID: PMC11434998 DOI: 10.3390/pharmaceutics16091120] [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: 06/20/2024] [Revised: 07/27/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Natural photosensitizers (PS) are compounds derived from nature, with photodynamic properties. Natural PSs have a similar action to that of commercial PSs, where cancer cell death occurs by necrosis, apoptosis, and autophagy through ROS generation. Natural PSs have garnered great interest over the last few decades because of their high biocompatibility and good photoactivity. Specific wavelengths could cause phytochemicals to produce harmful ROS for photodynamic therapy (PDT). However, natural PSs have some shortcomings, such as reduced solubility and lower uptake, making them less appropriate for PDT. Nanotechnology offers an opportunity to develop suitable carriers for various natural PSs for PDT applications. Various nanoparticles have been developed to improve the outcome with enhanced solubility, optical adsorption, and tumor targeting. Multidrug resistance (MDR) is a phenomenon in which tumor cells develop resistance to a wide range of structurally and functionally unrelated drugs. Over the last decade, several researchers have extensively studied the effect of natural PS-based photodynamic treatment (PDT) on MDR cells. Though the outcomes of clinical trials for natural PSs were inconclusive, significant advancement is still required before PSs can be used as a PDT agent for treating MDR tumors. This review addresses the increasing literature on MDR tumor progression and the efficacy of PDT, emphasizing the importance of developing new nano-based natural PSs in the fight against MDR that have the required features for an MDR tumor photosensitizing regimen.
Collapse
Affiliation(s)
- Jagadeesh Rajaram
- Department of Biochemistry and Molecular Medicine, National Dong Hwa University, Hualien 974, Taiwan;
| | - Lokesh Kumar Mende
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan;
| | - Yaswanth Kuthati
- Department of Anesthesiology, Cathy General Hospital, Taipei 106, Taiwan;
| |
Collapse
|
3
|
Cai X, Xu W, Ren C, Zhang L, Zhang C, Liu J, Yang C. Recent progress in quantitative analysis of self-assembled peptides. EXPLORATION (BEIJING, CHINA) 2024; 4:20230064. [PMID: 39175887 PMCID: PMC11335468 DOI: 10.1002/exp.20230064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/05/2023] [Indexed: 08/24/2024]
Abstract
Self-assembled peptides have been among the important biomaterials due to its excellent biocompatibility and diverse functions. Over the past decades, substantial progress and breakthroughs have been made in designing self-assembled peptides with multifaceted biomedical applications. The techniques for quantitative analysis, including imaging-based quantitative techniques, chromatographic technique and computational approach (molecular dynamics simulation), are becoming powerful tools for exploring the structure, properties, biomedical applications, and even supramolecular assembly processes of self-assembled peptides. However, a comprehensive review concerning these quantitative techniques remains scarce. In this review, recent progress in techniques for quantitative investigation of biostability, cellular uptake, biodistribution, self-assembly behaviors of self-assembled peptide etc., are summarized. Specific applications and roles of these techniques are highlighted in detail. Finally, challenges and outlook in this field are concluded. It is believed that this review will provide technical guidance for researchers in the field of peptide-based materials and pharmaceuticals, and facilitate related research for newcomers in this field.
Collapse
Affiliation(s)
- Xiaoyao Cai
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Wei Xu
- Department of PathologyCharacteristic Medical Center of Chinese People's Armed Police ForcesTianjinP. R. China
| | - Chunhua Ren
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Liping Zhang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Congrou Zhang
- Metabolomics and Analytics Center, Leiden Academic Centre of Drug ResearchLeiden UniversityLeidenThe Netherlands
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| | - Cuihong Yang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinP. R. China
| |
Collapse
|
4
|
Singh IR, Aggarwal N, Srivastava S, Panda JJ, Mishra J. Small Peptide-Based Nanodelivery Systems for Cancer Therapy and Diagnosis. J Pharmacol Exp Ther 2024; 390:30-44. [PMID: 37977815 DOI: 10.1124/jpet.123.001845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023] Open
Abstract
Developing nano-biomaterials with tunable topology, size, and surface characteristics has shown tremendously favorable benefits in various biologic and clinical applications. Among various nano-biomaterials, peptide-based drug delivery systems offer multiple merits over other synthetic systems due to their enhanced bio- and cytocompatibility and desirable biochemical and biophysical properties. Currently, around 100 peptide-based drugs are clinically available for numerous therapeutic purposes. In conjugation with chemotherapeutic moieties, peptides demonstrate a remarkable ability to reduce nonspecific drug effects by improving drug targetability at cancer sites. This review encompasses a wide-ranging role played by different peptide-based nanostructures in cancer theranostics. Section 1 introduces the rising concern about cancer as a disease and further describes peptide-based nanomaterials as biomedical agents to tackle the ailment. The subsequent section explores the mechanistic pathways behind the self-assembly of peptides to form hierarchically distinct assemblies. The crux of our review lies in an exhaustive exploration of the applications of various types of peptide-based nanostructures in cancer therapy and diagnosis. SIGNIFICANCE STATEMENT: Peptide-based drug delivery systems possess superior biocompatibility, biochemical, and biophysical properties compared to other synthetic alternatives. The development of these nano-biomaterials with customizable topology, size, and surface characteristics have shown promising outcomes in biomedical contexts. Peptides in conjunction with chemotherapeutic agents exhibit the ability to enhance drug targetability at cancer sites, reducing nonspecific drug effects. This comprehensive review emphasizes the pivotal role of diverse peptide-based nanostructures as cancer theranostics, elucidating their potential in revolutionizing cancer therapy and diagnosis.
Collapse
Affiliation(s)
- Imocha Rajkumar Singh
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Nidhi Aggarwal
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Swapnil Srivastava
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Jiban Jyoti Panda
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| | - Jibanananda Mishra
- Chemical Biology Unit, Institute of Nano Science and Technology, Mohali, India (I.R.S., N.A., S.S., J.J.P.) and School of Biosciences, RIMT University, Mandi Gobindgarh, India (J.M.)
| |
Collapse
|
5
|
Wang X, Zhao Y, Yan M, Liang X, Zhao N, Lu T. iRGD mediated pH-responsive mesoporous silica enhances drug accumulation in tumors. Eur J Pharm Sci 2024; 195:106725. [PMID: 38346565 DOI: 10.1016/j.ejps.2024.106725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/23/2024] [Accepted: 02/07/2024] [Indexed: 02/29/2024]
Abstract
The limited penetration of nanocarriers into tumors and the slow release of drugs from these carriers to tumor cells are significant challenges in cancer therapy. In this study, we developed a novel drug delivery carrier derived from mesoporous silica, dually modified with the tumor-homing cyclic peptide iRGD (CRGDKGPDC) and the pH-responsive polymer poly(2-ethyl-2-oxazoline) (PEOz) for treating triple-negative breast cancer. The carrier selectively bound to the αvβ3 integrin receptor, which is specifically expressed in MDA-MB-231 breast cancer cells and vessels. Subsequently, it penetrated deep into the tumor parenchyma through NRP-1 receptor-dependent internalization, with the drug-loaded particles releasing drugs rapidly in the acidic cytoplasmic environment. Results indicated that the drug release rate of PEOz-modified formulations was pH-dependent. Lysosomal escape experiments demonstrated that PEOz-modified particles efficiently escaped lysosomes to release drugs. In vitro cytotoxicity assays revealed that iRGD-functionalized particles were more cytotoxic to NRP-1-positive MDA-MB-231 cells compared to NRP-1-negative MCF-7 cells. Cellular uptake studies demonstrated that iRGD mediated enhanced endocytosis of nanoparticles into MDA-MB-231 cells. In vitro tumor cell spheroid penetration assays confirmed that the PEOz and iRGD dual-modified carrier facilitated deeper distribution of DOX in multicellular spheroids compared to free DOX. Moreover, in a nude mouse model of triple-negative breast cancer, the dual-modified drug-loaded carrier significantly inhibited tumor growth without inducing weight loss or liver and kidney damage. This dual-modified mesoporous silica presents a novel and promising delivery carrier for enhancing cancer treatment.
Collapse
Affiliation(s)
- Xiaoning Wang
- College of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi 710021, PR China; The School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China.
| | - Yangguang Zhao
- College of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi 710021, PR China
| | - Mengru Yan
- College of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi 710021, PR China
| | - Xiaoyan Liang
- College of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi 710021, PR China
| | - Ning Zhao
- College of Pharmacy, Xi'an Medical University, Xi'an, Shaanxi 710021, PR China
| | - Tingli Lu
- The School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, PR China
| |
Collapse
|
6
|
Shen X, Pan D, Gong Q, Gu Z, Luo K. Enhancing drug penetration in solid tumors via nanomedicine: Evaluation models, strategies and perspectives. Bioact Mater 2024; 32:445-472. [PMID: 37965242 PMCID: PMC10641097 DOI: 10.1016/j.bioactmat.2023.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 11/16/2023] Open
Abstract
Effective tumor treatment depends on optimizing drug penetration and accumulation in tumor tissue while minimizing systemic toxicity. Nanomedicine has emerged as a key solution that addresses the rapid clearance of free drugs, but achieving deep drug penetration into solid tumors remains elusive. This review discusses various strategies to enhance drug penetration, including manipulation of the tumor microenvironment, exploitation of both external and internal stimuli, pioneering nanocarrier surface engineering, and development of innovative tactics for active tumor penetration. One outstanding strategy is organelle-affinitive transfer, which exploits the unique properties of specific tumor cell organelles and heralds a potentially transformative approach to active transcellular transfer for deep tumor penetration. Rigorous models are essential to evaluate the efficacy of these strategies. The patient-derived xenograft (PDX) model is gaining traction as a bridge between laboratory discovery and clinical application. However, the journey from bench to bedside for nanomedicines is fraught with challenges. Future efforts should prioritize deepening our understanding of nanoparticle-tumor interactions, re-evaluating the EPR effect, and exploring novel nanoparticle transport mechanisms.
Collapse
Affiliation(s)
- Xiaoding Shen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, 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
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, 361021, China
| | - Zhongwei Gu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital Sichuan University, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, 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
|
7
|
Das P, N M, Singh N, Datta P. Supramolecular Nanostructures for the Delivery of Peptides in Cancer Therapy. J Pharmacol Exp Ther 2024; 388:67-80. [PMID: 37827700 DOI: 10.1124/jpet.123.001698] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Supramolecular nanostructured based delivery systems are emerging as a meaningful approach in the treatment of cancer, offering controlled drug release and improved therapeutic efficacy. The self-assembled structures can be small molecules, polymers, peptides, or proteins, which can be used and functionalized to achieve tailored release and target specific cells, tissues, or organs. These structures can improve the solubility and stability of drugs having low aqueous solubility by encapsulating and protecting them from degradation. Alongside, peptides as natural biomolecules have gained increasing attention as potential candidates in cancer treatment because of their biocompatibility, low cytotoxicity, and high specificity toward tumor cells. The amino acid sequences in peptide molecules are tunable, efficiently controlling the morphology of peptide-based self-assembled nanosystems and offering flexibility to form supramolecular nanostructures (SNs). It is evident from the current literature that the supramolecular nanostructures based delivery of peptide for cancer treatment hold great promise for future cancer therapy, offering potential strategies for personalized medicine with improved patient outcomes. SIGNIFICANCE STATEMENT: This review focuses on fundamentals and various drug delivery mechanisms based on SNs. Different SN approaches and recent literature reviews on peptide delivery are also presented to the readers.
Collapse
Affiliation(s)
- Priyanka Das
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | - Manasa N
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | - Nidhi Singh
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| | - Pallab Datta
- Polymer-Based Medical Devices and Complex Drug Delivery Systems Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, India
| |
Collapse
|
8
|
Zeng X, Yang M, Liu H, Zhang Z, Hu Y, Shi J, Wang ZH. Light-driven micro/nanomotors in biomedical applications. NANOSCALE 2023; 15:18550-18570. [PMID: 37962424 DOI: 10.1039/d3nr03760f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Nanotechnology brings hope for targeted drug delivery. However, most current drug delivery systems use passive delivery strategies with limited therapeutic efficiency. Over the past two decades, research on micro/nanomotors (MNMs) has flourished in the biomedical field. Compared with other driven methods, light-driven MNMs have the advantages of being reversible, simple to control, clean, and efficient. Under light irradiation, the MNMs can overcome several barriers in the body and show great potential in the treatment of various diseases, such as tumors, and gastrointestinal, cardiovascular and cerebrovascular diseases. Herein, the classification and mechanism of light-driven MNMs are introduced briefly. Subsequently, the applications of light-driven MNMs in overcoming physiological and pathological barriers in the past five years are highlighted. Finally, the future prospects and challenges of light-driven MNMs are discussed as well. This review will provide inspiration and direction for light-driven MNMs to overcome biological barriers in vivo and promote the clinical application of light-driven MNMs in the biomedical field.
Collapse
Affiliation(s)
- Xuejiao Zeng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Mingzhu Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Hua Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| | - Yurong Hu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| | - Zhi-Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, 450001, China
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou, 450001, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou, 450001, China
| |
Collapse
|
9
|
Jia L, Wang L, Song Y, Pang X, Zhao J. Exploring the sonodynamic effects of bacteriochlorophyll a. Front Bioeng Biotechnol 2023; 11:1186897. [PMID: 37251570 PMCID: PMC10213884 DOI: 10.3389/fbioe.2023.1186897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023] Open
Abstract
Objective: The purpose of this study was to investigate whether bacteriochlorophyll a (BCA) could be used as a potential diagnostic factor in near-infrared fluorescence (NIRF) imaging and in mediating sonodynamic antitumor effect. Methods: The UV spectrum and fluorescence spectra of bacteriochlorophyll a were measured. The IVIS Lumina imaging system was used to observe the fluorescence imaging of bacteriochlorophyll a. 9,10-Dimethylanthracene (DMA) reagent was used as a singlet oxygen sensor to detect singlet oxygen produced by bacteriochlorophyll a. LLC cells of mouse lung adenocarcinoma were selected as experimental subjects. Flow cytometry was used to detect the optimal uptake time of bacteriochlorophyll a in LLC cells. A laser confocal microscope was used to observe the binding of bacteriochlorophyll a to cells. The cell survival rate of each experimental group was detected by the CCK-8 method to detect the cytotoxicity of bacteriochlorophyll a. The effect of BCA-mediated sonodynamic therapy (SDT) on tumor cells was detected by the calcein acetoxymethyl ester/propidium iodide (CAM/PI) double staining method. 2,7-Dichlorodihydrofluorescein-diacetate (DCFH-DA) was used as the staining agent to evaluate and analyze intracellular reactive oxygen species (ROS) levels by fluorescence microscopy and flow cytometry (FCM). A confocal laser scanning microscope (CLSM) was used to observe the localization in the organelles of bacteriochlorophyll a. The IVIS Lumina imaging system was used to observe the fluorescence imaging of BCA in vitro. Results: Bacteriochlorophyll a-mediated SDT significantly increased cytotoxicity to LLC cells compared to other treatments, such as ultrasound (US) only, bacteriochlorophyll a only, and sham therapy. The CLSM observed bacteriochlorophyll a aggregation around the cell membrane and cytoplasm. FCM analysis and fluorescence microscopy showed that bacteriochlorophyll a-mediated SDT in LLC cells significantly inhibited cell growth and caused an obvious increase in intracellular ROS levels, and its fluorescence imaging function suggests that it can be a potential diagnostic factor. Conclusion: The results showed that bacteriochlorophyll a possesses good sonosensitivity and fluorescence imaging function. It can be effectively internalized in LLC cells, and bacteriochlorophyll a-mediated SDT is associated with ROS generation. This suggests that bacteriochlorophyll a can be used as a new type of sound sensitizer, and the bacteriochlorophyll a-mediated sonodynamic effect may be a potential treatment for lung cancer.
Collapse
Affiliation(s)
- Lanqi Jia
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Longhao Wang
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yiqiong Song
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Pang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jie Zhao
- Internet Medical and System Applications of National Engineering Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
10
|
Menilli L, Milani C, Reddi E, Moret F. Overview of Nanoparticle-Based Approaches for the Combination of Photodynamic Therapy (PDT) and Chemotherapy at the Preclinical Stage. Cancers (Basel) 2022; 14:cancers14184462. [PMID: 36139623 PMCID: PMC9496990 DOI: 10.3390/cancers14184462] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The present review represents the outstanding and promising recent literature reports (2017–2022) on nanoparticle-based formulations developed for anticancer therapy with photodynamic therapy (PDT), photosensitizers, and chemotherapeutics. Besides brief descriptions of chemotherapeutics’ classification and of PDT mechanisms and limitations, several examples of nanosystems endowed with different responsiveness (e.g., acidic pH and reactive oxygen species) and peculiarity (e.g., tumor oxygenation capacity, active tumor targeting, and biomimetic features) are described, and for each drug combination, in vitro and in vivo results on preclinical cancer models are reported. Abstract The widespread diffusion of photodynamic therapy (PDT) as a clinical treatment for solid tumors is mainly limited by the patient’s adverse reaction (skin photosensivity), insufficient light penetration in deeply seated neoplastic lesions, unfavorable photosensitizers (PSs) biodistribution, and photokilling efficiency due to PS aggregation in biological environments. Despite this, recent preclinical studies reported on successful combinatorial regimes of PSs with chemotherapeutics obtained through the drugs encapsulation in multifunctional nanometric delivery systems. The aim of the present review deals with the punctual description of several nanosystems designed not only with the objective of co-transporting a PS and a chemodrug for combination therapy, but also with the goal of improving the therapeutic efficacy by facing the main critical issues of both therapies (side effects, scarce tumor oxygenation and light penetration, premature drug clearance, unspecific biodistribution, etc.). Therefore, particular attention is paid to the description of bio-responsive drugs and nanoparticles (NPs), targeted nanosystems, biomimetic approaches, and upconverting NPs, including analyzing the therapeutic efficacy of the proposed photo-chemotherapeutic regimens in in vitro and in vivo cancer models.
Collapse
Affiliation(s)
- Luca Menilli
- Department of Biology, University of Padova, 35100 Padova, Italy
| | - Celeste Milani
- Department of Biology, University of Padova, 35100 Padova, Italy
- Institute of Organic Synthesis and Photoreactivity, ISOF-CNR, 40129 Bologna, Italy
| | - Elena Reddi
- Department of Biology, University of Padova, 35100 Padova, Italy
- Correspondence: (E.R.); (F.M.)
| | - Francesca Moret
- Department of Biology, University of Padova, 35100 Padova, Italy
- Correspondence: (E.R.); (F.M.)
| |
Collapse
|
11
|
Liu P, Fu Y, Wei F, Ma T, Ren J, Xie Z, Wang S, Zhu J, Zhang L, Tao J, Zhu J. Microneedle Patches with O 2 Propellant for Deeply and Fast Delivering Photosensitizers: Towards Improved Photodynamic Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202591. [PMID: 35839467 PMCID: PMC9443460 DOI: 10.1002/advs.202202591] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/29/2022] [Indexed: 05/31/2023]
Abstract
Photodynamic therapy (PDT) is an emerging technique for treating tumors. Especially, topical administration of photosensitizers (PSs) is more favorable for superficial tumor treatments with low systematic phototoxicity. Yet, ineffective migration of PSs to targeted tumor tissues and rapid consumption of O2 during PDT greatly limit their effects. Herein, PS-loaded microneedle (MN) patches with O2 propellant for a deeper and faster transdermal delivery of PS and improved PDT by embedding sodium percarbonate (SPC) into dissolving poly(vinyl pyrrolidone) MNs are presented. It is shown that SPC in the MNs can react with surrounding fluid to generate gaseous oxygen bubbles, forming vigorous fluid flows and thus greatly enhancing PS of chlorin e6 (Ce6) penetration in both hydrogel models and skin tissues. Reactive oxygen species (ROS) in hypoxic breast cancer cells (4T1 cells) are greatly increased by rapid penetration of PS and relief of hypoxia in vitro, and Ce6-loaded SPC MNs show an excellent cell-killing effect. Moreover, lower tumor growth rate and tumor mass after a 20-d treatment in tumor-bearing mice model verify the improved PDT in gaseous oxygen-droved delivery of PS. This study demonstrates a facile yet effective route of MN delivery of PSs for improved PDT in hypoxic tumor treatment.
Collapse
Affiliation(s)
- Pei Liu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materialsand State Key Laboratory of Materials Processing and Mold TechnologySchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST)Wuhan430074China
| | - Yangxue Fu
- Department of DermatologyUnion HospitalTongji Medical CollegeHUSTWuhan430022China
| | - Fulong Wei
- School of Energy and Power EngineeringHUSTWuhan430074China
| | - Teng Ma
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materialsand State Key Laboratory of Materials Processing and Mold TechnologySchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST)Wuhan430074China
| | - Jingli Ren
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materialsand State Key Laboratory of Materials Processing and Mold TechnologySchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST)Wuhan430074China
| | - Zhanjun Xie
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materialsand State Key Laboratory of Materials Processing and Mold TechnologySchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST)Wuhan430074China
| | - Shanzheng Wang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materialsand State Key Laboratory of Materials Processing and Mold TechnologySchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST)Wuhan430074China
| | - Jinjin Zhu
- Department of DermatologyUnion HospitalTongji Medical CollegeHUSTWuhan430022China
| | - Lianbin Zhang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materialsand State Key Laboratory of Materials Processing and Mold TechnologySchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST)Wuhan430074China
| | - Juan Tao
- Department of DermatologyUnion HospitalTongji Medical CollegeHUSTWuhan430022China
| | - Jintao Zhu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materialsand State Key Laboratory of Materials Processing and Mold TechnologySchool of Chemistry and Chemical EngineeringHuazhong University of Science and Technology (HUST)Wuhan430074China
| |
Collapse
|
12
|
Chen Z, Huang H, He S, Wang Y, Cai L, Xie Y. Progresses in Fluorescence Imaging Guidance for Bone and Soft Tissue Sarcoma Surgery. Front Oncol 2022; 12:879697. [PMID: 35860548 PMCID: PMC9289289 DOI: 10.3389/fonc.2022.879697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
R0 surgical resection is the preferred treatment for bone and soft tissue sarcoma. However, there is still a lack of precise technology that can visualize bone and soft tissue sarcoma during surgery to assist the surgeon in judging the tumor surgical boundary. Fluorescence imaging technology has been used in the diagnosis of cancer. It is a simple and essentially safe technique that takes no additional time during the operation. Intraoperative fluorescence imaging has potential application prospects in assisting the surgeons in judging the tumor boundary and improving the accuracy of surgical resection. This review mainly starts with clinical studies, animal experimentation, and newly designed probes of intraoperative fluorescence imaging of bone and soft tissue sarcoma, to appraise the application prospects of fluorescence imaging technology in bone and soft tissue sarcoma.
Collapse
|
13
|
Wang XJ, Cheng J, Zhang LY, Zhang JG. Self-assembling peptides-based nano-cargos for targeted chemotherapy and immunotherapy of tumors: recent developments, challenges, and future perspectives. Drug Deliv 2022; 29:1184-1200. [PMID: 35403517 PMCID: PMC9004497 DOI: 10.1080/10717544.2022.2058647] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Xue-Jun Wang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Jian Cheng
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
| | - Le-Yi Zhang
- Department of General Surgery, Chun’an First People’s Hospital (Zhejiang Provincial People’s Hospital Chun’an Branch), Hangzhou, China
| | - Jun-Gang Zhang
- General Surgery, Cancer Center, Department of Hepatobiliary and Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital of Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
14
|
Jing X, Hu H, Sun Y, Yu B, Cong H, Shen Y. The Intracellular and Extracellular Microenvironment of Tumor Site: The Trigger of Stimuli-Responsive Drug Delivery Systems. SMALL METHODS 2022; 6:e2101437. [PMID: 35048560 DOI: 10.1002/smtd.202101437] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The tumor microenvironment (TME), including intracellular and extracellular microenvironment, contains many biochemical indicators (such as acidity/alkalinity, oxygen content, and enzymatic activity) that are different from the normal physiological environment. These abnormal biochemical indicators can accelerate the heterogeneity of tumors, but on the other hand, they also provide opportunities for the design of intelligent drug delivery systems (DDSs). The TME-responsive DDSs have shown great potential in reducing the side effects of chemotherapy and improving the curative effect of tumors. In this review, the abnormal biochemical indicators of TME are introduced in detail from both the extracellular and intracellular aspects. In view of the various physiological barriers encountered during drug delivery, the strategy of constructing TME-responsive DDSs is discussed. By summarizing the typical research progress, the authors prospect the development of TME-responsive DDS in the future.
Collapse
Affiliation(s)
- Xiaodong Jing
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Yanzhen Sun
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
15
|
Multifunctional building elements for the construction of peptide drug conjugates. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
16
|
Chen B, Liu X, Li Y, Shan T, Bai L, Li C, Wang Y. iRGD Tumor-Penetrating Peptide-Modified Nano-Delivery System Based on a Marine Sulfated Polysaccharide for Enhanced Anti-Tumor Efficiency Against Breast Cancer. Int J Nanomedicine 2022; 17:617-633. [PMID: 35173433 PMCID: PMC8842734 DOI: 10.2147/ijn.s343902] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/13/2022] [Indexed: 12/16/2022] Open
Abstract
Background Breast cancer is a common malignancy in women. Conventional clinical therapies for breast cancer all display moderate clinical efficacies and limitations. It is urgent to explore the novel and combined therapeutic strategies for breast cancer to meet clinical demand. Methods An iRGD tumor-penetrating peptide-modified nano-delivery system (denoted as iRGD-PSS@PBAE@JQ1/ORI nanoparticles) based on a marine sulfated polysaccharide was developed by codelivery of JQ1 (BET inhibitor) and oridonin (ORI, bioactive diterpenoid derived from traditional Chinese medicine herb). The iRGD-PSS@PBAE@JQ1/ORI NPs, surface modified with iRGD peptide conjugated propylene glycol alginate sodium sulfate (iRGD-PSS). The antitumor efficacy was evaluated both in vitro and in vivo. Results The prepared iRGD-PSS@PBAE@JQ1/ORI NPs effectively enhanced the tumor targeting and cellular internalization of JQ1 and ORI. Thus, JQ1 exerted the reversal effect on immune tolerance by decreasing the expression of PD-L1, while ORI displayed multiple antitumor effects, such as antiproliferation, inhibition of intracellular ROS production and inhibition of lactic acid secretion. Conclusion Our data revealed that iRGD peptide could significantly improve the cellular internalization and tumor penetration of the nano-delivery system. The combination of JQ1 and ORI could exert synergistic antitumor activities. Taken together, this study provides a multifunctional nanotherapeutic system to enhance the anti-tumor efficiency against breast cancer.
Collapse
Affiliation(s)
- Bowei Chen
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Xiaohong Liu
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Yunan Li
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Tianhe Shan
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Liya Bai
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Chunyu Li
- Department of Integrated Traditional Chinese and Western Medicine, International Medical School, Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Yinsong Wang
- School of Pharmacy, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), Tianjin Medical University, Tianjin, 300070, People’s Republic of China
- Correspondence: Yinsong Wang; Chunyu Li, Email ;
| |
Collapse
|
17
|
Zhao C, Chen H, Wang F, Zhang X. Amphiphilic self-assembly peptides: Rational strategies to design and delivery for drugs in biomedical applications. Colloids Surf B Biointerfaces 2021; 208:112040. [PMID: 34425532 DOI: 10.1016/j.colsurfb.2021.112040] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/21/2021] [Accepted: 08/12/2021] [Indexed: 01/01/2023]
Abstract
Amphiphilic self-assembling peptides are widely used in tissue and cell engineering, antimicrobials, drug-delivery systems and other biomedical fields due to their good biocompatibility, functionality, flexibility of design and synthesis, and tremendous potential as delivery carriers for drugs. Currently, the design and study of amphipathic peptides by a bottom-up method to develop new biomedical materials have become a hot topic. However, defined rules have not been established for the design and development of self-assembled peptides. Therefore, the focus of this review is to summarize and provide several rational strategies for the design and study of amphiphilic self-assembly peptides. In addition, this paper also describes the types and general self-assembling mechanism of amphipathic peptides, and outlines their applications in the delivery of hydrophobic drugs, nucleic acid drugs, peptide drugs and vaccines. Amphiphilic self-assembled peptides are expected to exploit new functional materials for drug delivery and other applications.
Collapse
Affiliation(s)
- Chunqian Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China.
| | - Hongyuan Chen
- Department of General Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong University, Jinan, 250021, People's Republic of China.
| | - Fengshan Wang
- Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Quality Research and Evaluation of Carbohydrate-based Medicine, Institute of Biochemical and Biotechnological Drug, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China.
| | - Xinke Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, People's Republic of China.
| |
Collapse
|
18
|
Zhang M, Gao S, Yang D, Fang Y, Lin X, Jin X, Liu Y, Liu X, Su K, Shi K. Influencing factors and strategies of enhancing nanoparticles into tumors in vivo. Acta Pharm Sin B 2021; 11:2265-2285. [PMID: 34522587 PMCID: PMC8424218 DOI: 10.1016/j.apsb.2021.03.033] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/05/2021] [Accepted: 02/03/2021] [Indexed: 02/07/2023] Open
Abstract
The administration of nanoparticles (NPs) first faces the challenges of evading renal filtration and clearance of reticuloendothelial system (RES). After that, NPs infiltrate through the expanded endothelial space and penetrated the dense stroma of tumor microenvironment to tumor cells. As long as possible to prolong the time of NPs remaining in tumor tissue, NPs release active agent and induce pharmacological action. This review provides a comprehensive summary of the physical and chemical properties of NPs and the influence of various biological factors in tumor microenvironment, and discusses how to improve the final efficacy through adjusting the characteristics and structure of NPs. Perspectives and future directions are also provided.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kai Shi
- Corresponding author. Tel./fax: +86 24 43520557.
| |
Collapse
|
19
|
Zhou L, Pi W, Hao M, Li Y, An H, Li Q, Zhang P, Wen Y. An injectable and biodegradable nano-photothermal DNA hydrogel enhances penetration and efficacy of tumor therapy. Biomater Sci 2021; 9:4904-4921. [PMID: 34047319 DOI: 10.1039/d1bm00568e] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The biological barrier of solid tumors hinders deep penetration of nanomedicine, constraining anticancer treatment. Moreover, the inherent multidrug resistance (MDR) of cancer tissues may further limit the efficacy of anti-tumor nanomedicine. We synthesized highly permeable, photothermal, injectable, and positively charged biodegradable nucleic acid hydrogel (DNA-gel) nanoparticles to deliver cancer drugs. The nanoparticles are derived from photothermal materials containing black phosphorus quantum dots (BPQDs). The intra-tumoral BPQDs improve the sensitivity of tumor cells to photothermal therapy (PTT) and photodynamic treatment (PDT). Tumor cells take up the positively charged and controllable size DNA-gel nanoparticles, facilitating easy penetration and translocation of the particles across and within the cells. Mouse models demonstrated the anti-tumor activity of the DNA gel nanoparticles in vivo. In particular, the DNA gel nanoparticles enhanced clearance of both small and large tumor masses. Just 20 days after treatment, the tumor masses had been cleared. Compared to DOX chemotherapy alone, the DNA-gel treatment also significantly reduced drug resistance and improved the overall survival of mice with orthotopic breast tumors (83.3%, 78 d). Therefore, DNA gel nanoparticles are safe and efficient supplements for cancer therapy.
Collapse
Affiliation(s)
- Liping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Wei Pi
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing, 100044 China.
| | - Mingda Hao
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Yansheng Li
- Beijing Key Laboratory for Sensors, Beijing Information Science & Technology University, Beijing 100192, China
| | - Heng An
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| | - Qicheng Li
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing, 100044 China.
| | - Peixun Zhang
- Department of Orthopaedics and Trauma, Peking University People's Hospital, Beijing, 100044 China.
| | - Yongqiang Wen
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing 100083, China.
| |
Collapse
|
20
|
Xiao Q, Mai B, Nie Y, Yuan C, Xiang M, Shi Z, Wu J, Leung W, Xu C, Yao SQ, Wang P, Gao L. In Vitro and In Vivo Demonstration of Ultraefficient and Broad-Spectrum Antibacterial Agents for Photodynamic Antibacterial Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:11588-11596. [PMID: 33656316 DOI: 10.1021/acsami.0c20837] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Increasing threats from both pathogenic infections and antibiotic resistance highlight the pressing demand for nonantibiotic agents and alternative therapies. Herein, we report several new phenothiazinium-based derivatives, which could be readily synthesized via fragment-based assembly, which exhibited remarkable bactericidal activities both in vitro and in vivo. Importantly, in contrast to numerous clinically and preclinically used antibacterial photosensitizers, these compounds were able to eliminate various types of microorganisms, including Gram-(+) Staphylococcus aureus (S. aureus), Gram-(-) Escherichia coli, multidrug-resistant S. aureus, and their associated biofilms, at low drug and light dosages (e.g., 0.21 ng/mL in vitro and 1.63 ng/cm2 in vivo to eradicate S. aureus at 30 J/cm2). This study thus unveils the potential of these novel phenothiaziniums as potent antimicrobial agents for highly efficient photodynamic antibacterial chemotherapy.
Collapse
Affiliation(s)
- Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, P. R. China
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| | - Bingjie Mai
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, P. R. China
| | - Yichu Nie
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Chuang Yuan
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410000, P. R. China
- Department of Critical Care Medicine, The Second People's Hospital of Shenzhen & First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen 518035, P. R. China
| | - Menghua Xiang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Zihan Shi
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, P. R. China
| | - Juan Wu
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wingnang Leung
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chuanshan Xu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shao Q Yao
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| | - Pan Wang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, P. R. China
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen 518107, P. R. China
| |
Collapse
|
21
|
Caporale A, Adorinni S, Lamba D, Saviano M. Peptide-Protein Interactions: From Drug Design to Supramolecular Biomaterials. Molecules 2021; 26:1219. [PMID: 33668767 PMCID: PMC7956380 DOI: 10.3390/molecules26051219] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
The self-recognition and self-assembly of biomolecules are spontaneous processes that occur in Nature and allow the formation of ordered structures, at the nanoscale or even at the macroscale, under thermodynamic and kinetic equilibrium as a consequence of specific and local interactions. In particular, peptides and peptidomimetics play an elected role, as they may allow a rational approach to elucidate biological mechanisms to develop new drugs, biomaterials, catalysts, or semiconductors. The forces that rule self-recognition and self-assembly processes are weak interactions, such as hydrogen bonding, electrostatic attractions, and van der Waals forces, and they underlie the formation of the secondary structure (e.g., α-helix, β-sheet, polyproline II helix), which plays a key role in all biological processes. Here, we present recent and significant examples whereby design was successfully applied to attain the desired structural motifs toward function. These studies are important to understand the main interactions ruling the biological processes and the onset of many pathologies. The types of secondary structure adopted by peptides during self-assembly have a fundamental importance not only on the type of nano- or macro-structure formed but also on the properties of biomaterials, such as the types of interaction, encapsulation, non-covalent interaction, or covalent interaction, which are ultimately useful for applications in drug delivery.
Collapse
Affiliation(s)
- Andrea Caporale
- IC-CNR, c/o Area Science Park, S.S. 14 Km 163.5 Basovizza, 34149 Trieste, Italy;
| | - Simone Adorinni
- Dipartimento di Scienze Chimiche e Farmaceutiche di Università di Trieste, Via L. Giorgieri 1, 34127 Trieste, Italy;
| | - Doriano Lamba
- IC-CNR, c/o Area Science Park, S.S. 14 Km 163.5 Basovizza, 34149 Trieste, Italy;
- Istituto Nazionale Biostrutture e Biosistemi, Consorzio Interuniversitario, Viale delle Medaglie d’Oro 305, I-00136 Roma, Italy
| | - Michele Saviano
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche (IC-CNR), Via Giovanni Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
22
|
Wang Y, Luo S, Wu Y, Tang P, Liu J, Liu Z, Shen S, Ren H, Wu D. Highly Penetrable and On-Demand Oxygen Release with Tumor Activity Composite Nanosystem for Photothermal/Photodynamic Synergetic Therapy. ACS NANO 2020; 14:17046-17062. [PMID: 33290657 DOI: 10.1021/acsnano.0c06415] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
A deep penetrating and pH-responsive composite nanosystem was strategically developed to improve the efficacy of synergetic photothermal/photodynamic therapy (PTT/PDT) against hypoxic tumor. The designed nanosystem ([PHC]PP@HA NPs) was constructed by coloading hemoglobin (Hb) and chlorin e6 on polydopamine to build small-sized PHC NPs, which were encapsulated inside the polymer micelles (poly(ethylene glycol)-poly(ethylenimine)) and then capped with functionalized hyaluronic acid. The pH-responsive feature made [PHC]PP@HA NPs retain an initial size of ∼140 nm in blood circulation but rapidly release small PHC NPs (∼10 nm) with a high tumor-penetrating ability in the tumor microenvironment. The in vitro penetration experiment showed that the penetration depth of PHC NPs in the multicellular tumor spheroids exceeded 110 μm. The [PHC]PP@HA NPs exhibited excellent biocompatibility, deep tumor permeability, high photothermal conversion efficiency (47.09%), and low combination index (0.59) under hypoxic conditions. Notably, the nanosystem can freely adjust the release of oxygen and damaging PHC NPs in an on-demand manner on the basis of the feedback of tumor activity. This feedback tumor therapy significantly improved the synergistic effect of PTT/PDT and reduced its toxic side effects. The in vivo antitumor results showed that the tumor inhibition rate of [PHC]PP@HA NPs with an on-demand oxygen supply of Hb was ∼100%, which was much better than those of PTT alone and Hb-free nanoparticles ([PC]PP@HA NPs). Consequently, the [PHC]PP@HA NP-mediated PTT/PDT guided by feedback tumor therapy achieved an efficient tumor ablation with an extremely low tumor recurrence rate (8.3%) 60 d later, indicating the versatile potential of PTT/PDT.
Collapse
Affiliation(s)
- Ya Wang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Siyuan Luo
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Youshen Wu
- Department of Chemistry, School of Science, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Peng Tang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Jiajun Liu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zeying Liu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Shihong Shen
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Haozhe Ren
- Health Science Center, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, P. R. China
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
23
|
Yang H, Tong Z, Sun S, Mao Z. Enhancement of tumour penetration by nanomedicines through strategies based on transport processes and barriers. J Control Release 2020; 328:28-44. [PMID: 32858072 DOI: 10.1016/j.jconrel.2020.08.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Nanomedicines for antitumour therapy have been widely studied in recent decades, but only a few have been used in clinical applications. One of the most important reasons is the poor tumour permeability of the nanomedicines. In this three-part review, intravascular, transvascular and extravascular transport were introduced one by one according to their roles in the overall process of nanomedicine transport into tumours. Transportation obstacles, such as elevated interstitial fluid pressure (IFP), abnormal blood vessels, dense tumour extracellular matrix (ECM) and binding site barriers (BSB), were each discussed in the context of the respective transport processes. Furthermore, homologous resolution strategies were summarized on the basis of each transportation obstacle, such as the normalization of blood vessels, regulation of the tumour microenvironment (TME) and application of transformable nanoparticles. At the end of this review, we propose holistic, concrete, and innovative views for better tumour penetration of nanomedicines.
Collapse
Affiliation(s)
- Huang Yang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China.
| | - Zongrui Tong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Shichao Sun
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, PR China
| |
Collapse
|
24
|
Li Y, Chen M, Yao B, Lu X, Song B, Vasilatos SN, Zhang X, Ren X, Yao C, Bian W, Sun L. Dual pH/ROS-Responsive Nanoplatform with Deep Tumor Penetration and Self-Amplified Drug Release for Enhancing Tumor Chemotherapeutic Efficacy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2002188. [PMID: 32627387 DOI: 10.1002/smll.202002188] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/28/2020] [Indexed: 05/13/2023]
Abstract
Poor deep tumor penetration and incomplete intracellular drug release remain challenges for antitumor nanomedicine application in clinical settings. Herein, a nanomedicine (RLPA-NPs) is developed that can achieve prolonged blood circulation, deep tumor penetration, active-targeting of cancer cells, endosome/lysosome escape, and intracellular selectivity self-amplified drug release for effective drug delivery. The RLPA-NPs are constructed by encapsulation of a pH-sensitive polymer octadecylamine-poly(aspartate-1-(3-aminopropyl) imidazole) (OA-P(Asp-API)) and a ROS-generation agent, β-Lapachone (Lap), in micelles assembled by the tumor-penetration peptide internalizing RGD (iRGD)-modified ROS-responsive paclitaxel (PTX)-prodrug. iRGD could promote RLPA-NPs penetration into deep tumor tissue, and specific targeting to cancer cells. After internalization by cancer cells through receptor-mediated endocytosis, OA-P(Asp-API) can rapidly protonate in the endosome's acidic environment, resulting in RLPA-NPs escape from the endosome through the "proton sponge effect". At the same time, the RLPA-NPs micelle disassembles, releasing Lap and PTX-prodrug. Subsequently, the released Lap could generate ROS, consequently amplifying and accelerating PTX release to kill tumor cells. The in vitro and in vivo studies demonstrated that RLPA-NPs can significantly improve the therapeutic effect compared to control groups. Therefore, RLPA-NPs are a promising nanoplatform for overcoming multiple physiological and pathological barriers to enhance drug delivery.
Collapse
Affiliation(s)
- Yongfei Li
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Mie Chen
- Department of General Surgery, Pukou District Central Hospital, Pukou Branch of Jiangsu Province Hospital, Nanjing, 211899, China
| | - Bowen Yao
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Xun Lu
- Milken School of Public Health, George Washington University, Washington, DC, 20052, USA
| | - Boyang Song
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Shauna N Vasilatos
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xiang Zhang
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Shuyang People's Hospital, Suqian, 223600, China
| | - Xiaomei Ren
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Chang Yao
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Weihe Bian
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing, 210029, China
| | - Lizhu Sun
- Department of Oncology, The Affiliated Shuyang Hospital of Xuzhou Medical University, Shuyang People's Hospital, Suqian, 223600, China
| |
Collapse
|
25
|
Wu J, Sha J, Zhang C, Liu W, Zheng X, Wang P. Recent advances in theranostic agents based on natural products for photodynamic and sonodynamic therapy. VIEW 2020. [DOI: 10.1002/viw.20200090] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Jiasheng Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU‐CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
| | - Jie Sha
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU‐CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
| | - Chuangli Zhang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU‐CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
| | - Weimin Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU‐CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- School of Future Technology University of Chinese Academy of Sciences Beijing P.R. China
| | - Xiuli Zheng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU‐CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
| | - Pengfei Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU‐CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and Chemistry Chinese Academy of Sciences Beijing P.R. China
- School of Future Technology University of Chinese Academy of Sciences Beijing P.R. China
| |
Collapse
|
26
|
Zhang Y, Wang B, Zhao R, Zhang Q, Kong X. Multifunctional nanoparticles as photosensitizer delivery carriers for enhanced photodynamic cancer therapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 115:111099. [PMID: 32600703 DOI: 10.1016/j.msec.2020.111099] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/06/2019] [Accepted: 05/15/2020] [Indexed: 12/31/2022]
Abstract
Photodynamic therapy (PDT) is an emerging cancer treatment combining light, oxygen, and a photosensitizer (PS) to produce highly cytotoxic reactive oxygen species that cause cancer cell death. However, most PSs are hydrophobic molecules that have poor water solubility and cannot target tumor tissues, causing damage to normal tissues and cells during PDT. Thus, there is a substantial demand for the development of nanocarrier systems to achieve targeted delivery of PSs into tumor tissues and cells. This review summarizes the research progress in PS delivery systems for PDT treatment of tumors and focuses on the recent design and development of multifunctional nanoparticles as PS delivery carriers for enhanced PDT. These multifunctional nanoparticles possess unique properties, including tunable particle size, changeable shape, stimuli-responsive PS activation, controlled PS release, and hierarchical targeting capability. These properties can increase tumor accumulation, penetration, and cellular internalization of nanoparticles to achieve PS activation and/or release in cancer cells for enhanced PDT. Finally, recent developments in multifunctional nanoparticles for tumor-targeted PS delivery and their future prospects in PDT are discussed.
Collapse
Affiliation(s)
- Yonghe Zhang
- Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Beilei Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Ruibo Zhao
- Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - Quan Zhang
- Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, China.
| | - Xiangdong Kong
- Key Laboratory of Advanced Textile Materials and Manufacturing Technology of Ministry of Education, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China; Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
27
|
Feger G, Angelov B, Angelova A. Prediction of Amphiphilic Cell-Penetrating Peptide Building Blocks from Protein-Derived Amino Acid Sequences for Engineering of Drug Delivery Nanoassemblies. J Phys Chem B 2020; 124:4069-4078. [PMID: 32337991 DOI: 10.1021/acs.jpcb.0c01618] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Amphiphilic molecules, forming self-assembled nanoarchitectures, are typically composed of hydrophobic and hydrophilic domains. Peptide amphiphiles can be designed from two, three, or four building blocks imparting novel structural and functional properties and affinities for interaction with cellular membranes or intracellular organelles. Here we present a combined numerical approach to design amphiphilic peptide scaffolds that are derived from the human nuclear Ki-67 protein. Ki-67 acts, like a biosurfactant, as a steric and electrostatic charge barrier against the collapse of mitotic chromosomes. The proposed predictive design of new Ki-67 protein-derived amphiphilic amino acid sequences exploits the computational outcomes of a set of web-accessible predictors, which are based on machine learning methods. The ensemble of such artificial intelligence algorithms, involving support vector machine (SVM), random forest (RF) classifiers, and neural networks (NN), enables the nanoengineering of a broad range of innovative peptide materials for therapeutic delivery in various applications. Amphiphilic cell-penetrating peptides (CPP), derived from natural protein sequences, may spontaneously form self-assembled nanocarriers characterized by enhanced cellular uptake. Thanks to their inherent low immunogenicity, they may enable the safe delivery of therapeutic molecules across the biological barriers.
Collapse
Affiliation(s)
- Guillaume Feger
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay UMR8612, F-92296 Châtenay-Malabry, France
| | - Borislav Angelov
- Institute of Physics, ELI Beamlines, Academy of Sciences of the Czech Republic, Na Slovance 2, CZ-18221 Prague, Czech Republic
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay UMR8612, F-92296 Châtenay-Malabry, France
| |
Collapse
|
28
|
Xiao Q, Lin H, Wu J, Pang X, Zhou Q, Jiang Y, Wang P, Leung W, Lee H, Jiang S, Yao SQ, Gao L, Liu G, Xu C. Pyridine-Embedded Phenothiazinium Dyes as Lysosome-Targeted Photosensitizers for Highly Efficient Photodynamic Antitumor Therapy. J Med Chem 2020; 63:4896-4907. [DOI: 10.1021/acs.jmedchem.0c00280] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Qicai Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Huirong Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Centre for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Juan Wu
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xin Pang
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Centre for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Quanming Zhou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Yue Jiang
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Pan Wang
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wingnang Leung
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hungkay Lee
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Sheng Jiang
- State Key Laboratory of Natural Medicines, Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shao Q. Yao
- Department of Chemistry, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| | - Liqian Gao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Centre for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Chuanshan Xu
- Key Laboratory of Molecular Target and Clinical Pharmacology, School of Pharmaceutical Sciences & Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
29
|
Otieno W, Liu C, Deng H, Li J, Zeng X, Ji Y. Hypocrellin B-Mediated Photodynamic Inactivation of Gram-Positive Antibiotic-Resistant Bacteria: An In Vitro Study. PHOTOBIOMODULATION PHOTOMEDICINE AND LASER SURGERY 2020; 38:36-42. [DOI: 10.1089/photob.2019.4656] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Woodvine Otieno
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Chengcheng Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Hong Deng
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Photochemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P.R. China
| | - Jiao Li
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| | - Xiaoyan Zeng
- The First Affiliated Hospital of College of Medicine, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Yanhong Ji
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, P.R. China
| |
Collapse
|
30
|
Kim H, Kim I, Hwang JH, Park J, Ahn H, Han EH, Lee E. Glutathione-adaptive peptide amphiphile vesicles rationally designed using positionable disulfide-bridges for effective drug transport. Polym Chem 2020. [DOI: 10.1039/d0py00504e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The drug loading/releasing capability of GSH-responsive nanovesicles self-assembled from peptide amphiphiles was controlled by varying the location and number of disulfide-linkages in the peptide for the selective drug-release into tumor cells.
Collapse
Affiliation(s)
- Hayeon Kim
- School of Materials Science and Engineering
- Gwangju Institute of Science and Technology (GIST)
- Gwangju 61005
- Republic of Korea
| | - Inhye Kim
- School of Materials Science and Engineering
- Gwangju Institute of Science and Technology (GIST)
- Gwangju 61005
- Republic of Korea
| | - Jun Ho Hwang
- School of Materials Science and Engineering
- Gwangju Institute of Science and Technology (GIST)
- Gwangju 61005
- Republic of Korea
| | - Jaehyun Park
- School of Materials Science and Engineering
- Gwangju Institute of Science and Technology (GIST)
- Gwangju 61005
- Republic of Korea
| | - Hyungju Ahn
- Pohang Accelerator Laboratory
- Pohang University of Science and Technology
- Pohang 37673
- Republic of Korea
| | - Eun Hee Han
- Division of Bioconvergence Analysis
- Korea Basic Science Institute (KBSI)
- Cheongju 28119
- Republic of Korea
| | - Eunji Lee
- School of Materials Science and Engineering
- Gwangju Institute of Science and Technology (GIST)
- Gwangju 61005
- Republic of Korea
| |
Collapse
|
31
|
Wang H, Jia Q, Liu W, Nan F, Zheng X, Ding Y, Ren H, Wu J, Ge J. Hypocrellin Derivative‐Loaded Calcium Phosphate Nanorods as NIR Light‐Triggered Phototheranostic Agents with Enhanced Tumor Accumulation for Cancer Therapy. ChemMedChem 2019; 15:177-181. [DOI: 10.1002/cmdc.201900512] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/14/2019] [Indexed: 11/09/2022]
Affiliation(s)
- Hongyi Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
| | - Qingyan Jia
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
- Xi'an Institute of Flexible Electronics (IFE)&Xi'an Institute of Biomedical Materials and Engineering (IBME)Northwestern Polytechnical University (NPU) 127 West Youyi Road Xi'an 710072 China
| | - Weimin Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
- School of Future TechnologyUniversity of Chinese Academy of Sciences Beijing 100049 China
| | - Fuchun Nan
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
- School of Future TechnologyUniversity of Chinese Academy of Sciences Beijing 100049 China
| | - Xiuli Zheng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
- School of Future TechnologyUniversity of Chinese Academy of Sciences Beijing 100049 China
| | - Ying Ding
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
- School of Future TechnologyUniversity of Chinese Academy of Sciences Beijing 100049 China
| | - Haohui Ren
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
| | - Jiasheng Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
| | - Jiechao Ge
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices Technical Institute of Physics and ChemistryChinese Academy of Sciences Beijing 100190 China
- School of Future TechnologyUniversity of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
32
|
Membrane intercalation-enhanced photodynamic inactivation of bacteria by a metallacycle and TAT-decorated virus coat protein. Proc Natl Acad Sci U S A 2019; 116:23437-23443. [PMID: 31685638 DOI: 10.1073/pnas.1911869116] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Antibiotic resistance has become one of the major threats to global health. Photodynamic inactivation (PDI) develops little antibiotic resistance; thus, it becomes a promising strategy in the control of bacterial infection. During a PDI process, light-induced reactive oxygen species (ROS) damage the membrane components, leading to the membrane rupture and bacteria death. Due to the short half-life and reaction radius of ROS, achieving the cell-membrane intercalation of photosensitizers is a key challenge for PDI of bacteria. In this work, a tetraphenylethylene-based discrete organoplatinum(II) metallacycle (1) acts as a photosensitizer with aggregation-induced emission. It self-assembles with a transacting activator of transduction (TAT) peptide-decorated virus coat protein (2) through electrostatic interactions. This assembly (3) exhibits both ROS generation and strong membrane-intercalating ability, resulting in significantly enhanced PDI efficiency against bacteria. By intercalating in the bacterial cell membrane or entering the bacteria, assembly 3 decreases the survival rate of gram-negative Escherichia coli to nearly zero and that of gram-positive Staphylococcus aureus to ∼30% upon light irradiation. This study has wide implications from the generation of multifunctional nanomaterials to the control of bacterial infection, especially for gram-negative bacteria.
Collapse
|
33
|
Wang G, Liu J, Zhu L, Guo Y, Yang L. Silver sulfide nanoparticles for photodynamic therapy of human lymphoma cells via disruption of energy metabolism. RSC Adv 2019; 9:29936-29941. [PMID: 35531500 PMCID: PMC9072148 DOI: 10.1039/c9ra05432d] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/09/2019] [Indexed: 01/11/2023] Open
Abstract
Recently, studies on the application of light-responsive semiconductor nanomaterials for photodynamic therapy (PDT) of non-solid tumors have attracted tremendous attention. Herein, 6.98 nm Ag2S nanoparticles (Ag2S NPs) with excellent aqueous dispersibility, stability, and biocompatibility were synthesized by a facile strategy without any post-modification. In vitro studies indicated that Ag2S NPs could significantly inhibit the growth of human lymphoma cells (Raji cells) compared with hepatoma carcinoma cells (Hep G2 cells) under light irradiation. Further studies revealed that Ag2S NPs could specifically induce the accumulation of intracellular reactive oxidative species in Raji cells under light irradiation, and induce significant disruption of energy metabolism. This finding provides inspiration for the potential application of Ag2S semiconductor nanoparticles as a photosensitizer to significantly and specifically treat human lymphoma through PDT. Ag2S/BSA hybrid nanoparticles were prepared and studied for their ability to inhibit the growth of human lymphoma cells under light irradiation, via inducing the accumulation of intracellular reactive oxidative species to disrupt energy metabolism.![]()
Collapse
Affiliation(s)
- Ge Wang
- Henan Key Laboratory of Green Chemical Media and Reactions, School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang Henan 453007 P. R. China .,School of Basic Medical Sciences, Xinxiang Medical University Xinxiang Henan 453003 P. R. China
| | - Jing Liu
- Henan Key Laboratory of Green Chemical Media and Reactions, School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang Henan 453007 P. R. China
| | - Lin Zhu
- Henan Key Laboratory of Green Chemical Media and Reactions, School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang Henan 453007 P. R. China
| | - Yuming Guo
- Henan Key Laboratory of Green Chemical Media and Reactions, School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang Henan 453007 P. R. China
| | - Lin Yang
- Henan Key Laboratory of Green Chemical Media and Reactions, School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang Henan 453007 P. R. China
| |
Collapse
|
34
|
Wang S, Yu G, Wang Z, Jacobson O, Lin L, Yang W, Deng H, He Z, Liu Y, Chen Z, Chen X. Enhanced Antitumor Efficacy by a Cascade of Reactive Oxygen Species Generation and Drug Release. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201908997] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Sheng Wang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging The Third Affiliated Hospital of Guangzhou Medical University The Liwan Hospital of the Third Affiliated Hospital of, Guangzhou Medical University Guangzhou Guangdong 510000 China
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Li‐Sen Lin
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Weijing Yang
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Hongzhang Deng
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Zhimei He
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Yuan Liu
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| | - Zhi‐Yi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging The Third Affiliated Hospital of Guangzhou Medical University The Liwan Hospital of the Third Affiliated Hospital of, Guangzhou Medical University Guangzhou Guangdong 510000 China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine National Institute of Biomedical Imaging and Bioengineering National Institutes of Health Bethesda MD 20892 USA
| |
Collapse
|
35
|
Wang S, Yu G, Wang Z, Jacobson O, Lin LS, Yang W, Deng H, He Z, Liu Y, Chen ZY, Chen X. Enhanced Antitumor Efficacy by a Cascade of Reactive Oxygen Species Generation and Drug Release. Angew Chem Int Ed Engl 2019; 58:14758-14763. [PMID: 31429173 DOI: 10.1002/anie.201908997] [Citation(s) in RCA: 178] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) can be used not only as a therapeutic agent for chemodynamic therapy (CDT), but also as a stimulus to activate release of antitumor drugs, achieving enhanced efficacy through the combination of CDT and chemotherapy. Here we report a pH/ROS dual-responsive nanomedicine consisting of β-lapachone (Lap), a pH-responsive polymer, and a ROS-responsive polyprodrug. In the intracellular acidic environment, the nanomedicine can realize pH-triggered disassembly. The released Lap can efficiently generate hydrogen peroxide, which will be further converted into highly toxic hydroxyl radicals via the Fenton reaction. Subsequently, through ROS-induced cleavage of thioketal linker, doxorubicin is released from the polyprodrug. In vivo results indicate that the cascade of ROS generation and antitumor-drug release can effectively inhibit tumor growth. This design of nanomedicine with cascade reactions offers a promising strategy to enhance antitumor efficacy.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China.,Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Li-Sen Lin
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weijing Yang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hongzhang Deng
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zhimei He
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuan Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zhi-Yi Chen
- Department of Ultrasound Medicine, Laboratory of Ultrasound Molecular Imaging, The Third Affiliated Hospital of Guangzhou Medical University, The Liwan Hospital of the Third Affiliated Hospital of, Guangzhou Medical University, Guangzhou, Guangdong, 510000, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
36
|
Feng X, Dixon H, Glen‐Ravenhill H, Karaosmanoglu S, Li Q, Yan L, Chen X. Smart Nanotechnologies to Target Tumor with Deep Penetration Depth for Efficient Cancer Treatment and Imaging. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Xue Feng
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Hannah Dixon
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Harriet Glen‐Ravenhill
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Sena Karaosmanoglu
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Quan Li
- School of EngineeringInstitute for Energy SystemsThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
| | - Li Yan
- Monash Institute of Pharmaceutical SciencesMonash University Parkville Victoria 3052 Australia
| | - Xianfeng Chen
- School of EngineeringInstitute for BioengineeringThe University of Edinburgh King's Buildings, Mayfield Road Edinburgh EH9 3JL UK
- Translational Medicine CenterThe Second Affiliated HospitalGuangzhou Medical University Guangzhou 510182 P. R. China
| |
Collapse
|
37
|
Kim J, Narayana A, Patel S, Sahay G. Advances in intracellular delivery through supramolecular self-assembly of oligonucleotides and peptides. Theranostics 2019; 9:3191-3212. [PMID: 31244949 PMCID: PMC6567962 DOI: 10.7150/thno.33921] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/09/2019] [Indexed: 12/15/2022] Open
Abstract
Cells utilize natural supramolecular assemblies to maintain homeostasis and biological functions. Naturally inspired modular assembly of biomaterials are now being exploited for understanding or manipulating cell biology for treatment, diagnosis, and detection of diseases. Supramolecular biomaterials, in particular peptides and oligonucleotides, can be precisely tuned to have diverse structural, mechanical, physicochemical and biological properties. These merits of oligonucleotides and peptides as building blocks have given rise to the evolution of numerous nucleic acid- and peptide-based self-assembling nanomaterials for various medical applications, including drug delivery, tissue engineering, regenerative medicine, and immunotherapy. In this review, we provide an extensive overview of the intracellular delivery approaches using supramolecular self-assembly of DNA, RNA, and peptides. Furthermore, we discuss the current challenges related to subcellular delivery and provide future perspectives of the application of supramolecular biomaterials for intracellular delivery in theranostics.
Collapse
Affiliation(s)
- Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
| | - Ashwanikumar Narayana
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
| | - Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
- Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health Science University, Portland, OR
| |
Collapse
|
38
|
Tesauro D, Accardo A, Diaferia C, Milano V, Guillon J, Ronga L, Rossi F. Peptide-Based Drug-Delivery Systems in Biotechnological Applications: Recent Advances and Perspectives. Molecules 2019; 24:E351. [PMID: 30669445 PMCID: PMC6359574 DOI: 10.3390/molecules24020351] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/09/2019] [Accepted: 01/18/2019] [Indexed: 12/30/2022] Open
Abstract
Peptides of natural and synthetic sources are compounds operating in a wide range of biological interactions. They play a key role in biotechnological applications as both therapeutic and diagnostic tools. They are easily synthesized thanks to solid-phase peptide devices where the amino acid sequence can be exactly selected at molecular levels, by tuning the basic units. Recently, peptides achieved resounding success in drug delivery and in nanomedicine smart applications. These applications are the most significant challenge of recent decades: they can selectively deliver drugs to only pathological tissues whilst saving the other districts of the body. This specific feature allows a reduction in the drug side effects and increases the drug efficacy. In this context, peptide-based aggregates present many advantages, including biocompatibility, high drug loading capacities, chemical diversity, specific targeting, and stimuli responsive drug delivery. A dual behavior is observed: on the one hand they can fulfill a structural and bioactive role. In this review, we focus on the design and the characterization of drug delivery systems using peptide-based carriers; moreover, we will also highlight the peptide ability to self-assemble and to actively address nanosystems toward specific targets.
Collapse
Affiliation(s)
- Diego Tesauro
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| | - Antonella Accardo
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| | - Carlo Diaferia
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| | - Vittoria Milano
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
- ARNA, INSERM U1212/UMR CNRS 5320, UFR des Sciences Pharmaceutiques, Université de Bordeaux, F-33000 Bordeaux, France.
| | - Jean Guillon
- ARNA, INSERM U1212/UMR CNRS 5320, UFR des Sciences Pharmaceutiques, Université de Bordeaux, F-33000 Bordeaux, France.
| | - Luisa Ronga
- Institute of Analytical Sciences, IPREM, UMR 5254, CNRS-University of Pau, 64000 Pau, France.
| | - Filomena Rossi
- Department of Pharmacy and CIRPeB, Università Federico II, 80134 Naples, Italy.
| |
Collapse
|
39
|
Chen Y, Fu Y, Li X, Chen H, Wang Z, Zhang H. Peptide-functionalized NaGdF4 nanoparticles for tumor-targeted magnetic resonance imaging and effective therapy. RSC Adv 2019; 9:17093-17100. [PMID: 35519897 PMCID: PMC9064544 DOI: 10.1039/c9ra02135c] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/21/2019] [Indexed: 01/05/2023] Open
Abstract
Metallic nanoparticles showed potent efficacy for diagnosis and therapy of cancer, but their clinical applications are limited by their poor tumor-targeting ability. Herein, peptide-functionalized 9 nm NaGdF4 nanoparticles (termed as, NaGdF4@bp-peptide NPs) have been synthesized through the Gd–phosphate coordination reaction of the spherical NaGdF4 nanoparticles with phosphopeptides (sequence: KLAKLAKKLAKLAKG(p-S)GAKRGARSTA, p-S means phosphorylated serine) including a p32 protein binding motif incorporating a cell-penetrating function, and a proapoptotic domain. The NaGdF4@bp-peptide NPs are ready to be efficiently internalized by cancer cells; they show a much higher cytotoxicity in MCF-7 breast cancer cells than the casein phosphopeptide (CPP) modified NaGdF4 nanoparticles (termed as, NaGdF4@CPP NPs). Using mouse-bearing MCF-7 breast cancer as a model system, the in vivo experimental results demonstrate that NaGdF4@bp-peptide NPs have integration of T1-weighted magnetic resonance imaging (MRI) contrast and tumor-targeting functionalities, and are able to suppress tumor growth without causing systemic toxicity. NaGdF4@bp-peptide nanoparticles have been used as a T1-weighted MR contrast agent with active-tumor targeting and antitumor ability.![]()
Collapse
Affiliation(s)
- Yixin Chen
- Department of Radiology
- The First Hospital of Jilin University
- Changchun 130021
- P. R. China
| | - Yu Fu
- Department of Radiology
- The First Hospital of Jilin University
- Changchun 130021
- P. R. China
| | - Xiaodong Li
- Department of Radiology
- The First Hospital of Jilin University
- Changchun 130021
- P. R. China
| | - Hongda Chen
- State Key Laboratory of Electroanalytical Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- P. R. China
| | - Zhenxin Wang
- State Key Laboratory of Electroanalytical Chemistry
- Changchun Institute of Applied Chemistry
- Chinese Academy of Sciences
- Changchun
- P. R. China
| | - Huimao Zhang
- Department of Radiology
- The First Hospital of Jilin University
- Changchun 130021
- P. R. China
| |
Collapse
|