1
|
Peng L, Gao Z, Liang Y, Guo X, Zhang Q, Cui D. Nanoparticle-based drug delivery systems: opportunities and challenges in the treatment of esophageal squamous cell carcinoma (ESCC). NANOSCALE 2025; 17:8270-8288. [PMID: 40052671 DOI: 10.1039/d4nr05114a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is an aggressive malignancy characterized by limited treatment options and poor prognosis. Nanoparticle-based drug delivery systems have emerged as a promising strategy to enhance cancer therapy efficacy by improving drug targeting, reducing toxicity, and enabling multifunctional applications. This review highlights some key types of nanoparticles, including liposomes, polymeric nanoparticles, metallic nanoparticles, dendrimers, and quantum dots, which could effectively improve the delivery of various drugs used in chemotherapy, radiotherapy, and immunotherapy, offering more precise and effective treatment options. With the ability to improve drug stability and overcome biological barriers, nanoparticle-based systems represent a transformative strategy for ESCC treatment. Despite some challenges, such as biocompatibility and scalability, the future of nanoparticle-based drug delivery holds great promise, particularly in the development of personalized nanomedicine and novel therapeutic approaches targeting the tumor microenvironment. With ongoing advancements, nanoparticle-based drug delivery systems hold immense potential to revolutionize ESCC treatment and improve patient outcomes.
Collapse
Affiliation(s)
- Linjia Peng
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Zixuan Gao
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Yanfeng Liang
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Xiaonan Guo
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Qiuli Zhang
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Daxiang Cui
- The First Afffliated Hospital of Henan University, N. Jinming Ave, Kaifeng, 475004, China
| |
Collapse
|
2
|
Jia W, Wu Y, Xie Y, Yu M, Chen Y. Advanced Polymeric Nanoparticles for Cancer Immunotherapy: Materials Engineering, Immunotherapeutic Mechanism and Clinical Translation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413603. [PMID: 39797474 DOI: 10.1002/adma.202413603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Indexed: 01/13/2025]
Abstract
Cancer immunotherapy, which leverages immune system components to treat malignancies, has emerged as a cornerstone of contemporary therapeutic strategies. Yet, critical concerns about the efficacy and safety of cancer immunotherapies remain formidable. Nanotechnology, especially polymeric nanoparticles (PNPs), offers unparalleled flexibility in manipulation-from the chemical composition and physical properties to the precision control of nanoassemblies. PNPs provide an optimal platform to amplify the potency and minimize systematic toxicity in a broad spectrum of immunotherapeutic modalities. In this comprehensive review, the basics of polymer chemistry, and state-of-the-art designs of PNPs from a physicochemical standpoint for cancer immunotherapy, encompassing therapeutic cancer vaccines, in situ vaccination, adoptive T-cell therapies, tumor-infiltrating immune cell-targeted therapies, therapeutic antibodies, and cytokine therapies are delineated. Each immunotherapy necessitates distinctively tailored design strategies in polymeric nanoplatforms. The extensive applications of PNPs, and investigation of their mechanisms of action for enhanced efficacy are particularly focused on. The safety profiles of PNPs and clinical research progress are discussed. Additionally, forthcoming developments and emergent trends of polymeric nano-immunotherapeutics poised to transform cancer treatment paradigms into clinics are explored.
Collapse
Affiliation(s)
- Wencong Jia
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Ye Wu
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai China, 200444, China
| | - Meihua Yu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
- Shanghai Institute of Materdicine, Shanghai, 200051, China
| |
Collapse
|
3
|
Shen Y, Sun Y, Liang Y, Xu X, Su R, Wang Y, Qi W. Full-color peptide-based fluorescent nanomaterials assembled under the control of amino acid doping. NANOSCALE HORIZONS 2024; 10:158-164. [PMID: 39498619 DOI: 10.1039/d4nh00400k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/06/2024]
Abstract
Peptide-based biofluorescents are of great interest due to their controllability and biocompatibility, as well as their potential applications in biomedical imaging and biosensing. Here, we present a simple approach to synthesizing full-color fluorescent nanomaterials with broad-spectrum fluorescence emissions, high optical stability, and long fluorescence lifetimes. By doping amino acids during the enzyme-catalyzed oxidative self-assembly of tyrosine-based peptides, we can precisely control the intermolecular interactions to obtain nanoparticles with fluorescence emission at different wavelengths. The synthesized peptide-based fluorescent nanomaterials with excellent biocompatibility and stable near-infrared fluorescence emission were shown to have potential for bioimaging applications. This research provides new ideas for the development of new bioluminescent materials that are cost-effective, environmentally friendly, and safe for biomedical use.
Collapse
Affiliation(s)
- Yuhe Shen
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Yulin Sun
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Yaoyu Liang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Xiaojian Xu
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- Collaborative Innovation Centre of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yuefei Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Wei Qi
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China.
- Collaborative Innovation Centre of Chemical Science and Engineering (Tianjin), Tianjin 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin 300072, P. R. China
| |
Collapse
|
4
|
Yuan C, Fan W, Zhou P, Xing R, Cao S, Yan X. High-entropy non-covalent cyclic peptide glass. NATURE NANOTECHNOLOGY 2024; 19:1840-1848. [PMID: 39187585 DOI: 10.1038/s41565-024-01766-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 07/19/2024] [Indexed: 08/28/2024]
Abstract
Biomolecule-based non-covalent glasses are biocompatible and biodegradable, and offer a sustainable alternative to conventional glass. Cyclic peptides (CPs) can serve as promising glass formers owing to their structural rigidity and resistance to enzymatic degradation. However, their potent crystallization tendency hinders their potential in glass construction. Here we engineered a series of CP glasses with tunable glass transition behaviours by modulating the conformational complexity of CP clusters. By incorporating multicomponent CPs, the formation of high-entropy CP glass is facilitated, which-in turn-inhibits the crystallization of individual CPs. The high-entropy CP glass demonstrates enhanced mechanical properties and enzyme tolerance compared with individual CP glass and a unique biorecycling capability that is unattainable by traditional glasses. These findings provide a promising paradigm for the design and development of stable non-covalent glasses based on naturally derived biomolecules, and advance their application in pharmaceutical formulations and smart functional materials.
Collapse
Affiliation(s)
- Chengqian Yuan
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Wei Fan
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Peng Zhou
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Ruirui Xing
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Shuai Cao
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
| | - Xuehai Yan
- State Key Laboratory of Biochemical Engineering, Key Laboratory of Biopharmaceutical Preparation and Delivery, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China.
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China.
- Center for Mesoscience, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
5
|
Yan R, Zhang N, Liu W, Hu X, Wang W, Tang Y, Wang S, Wang X, Sheng Q. Novel Eu-dipeptide assemblies for a fluorescence sensing strategy to ultrasensitive determine trace sulfamethazine. Food Chem 2024; 448:139089. [PMID: 38518446 DOI: 10.1016/j.foodchem.2024.139089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/11/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
Self-assembled Eu-dipeptide (tryptophan-phenylalanine) microparticles with multi-emission fluorescence was prepared and modified with a single-stranded DNA corresponding to the sulfamethazine (SMZ) adapter (Eu-PMPs@cDNA). Aptamer-functionalized magnetic Fe3O4 (MNPs@aptamer) was used to specifically bind the target SMZ. Using Eu-PMPs@cDNA as fluorescent signal probe and MNPs@aptamer as catcher, a noncompetitive fluorescence sensing strategy was developed for determination of SMZ with good sensitivity, accuracy, selectivity, and stability. Under the optimized conditions, fluorescence increases linearly in the 0-20 ng/mL SMZ concentration range, and the detection limit is 0.014 ng/mL. The fluorescence sensing method was applied to analysis of water and fish muscle samples, and recoveries ranged from 81.78 to 119.46 % with relative standard deviations below 4.2 %. This study offered a reliable and sensitive fluorescence sensing strategy for SMZ determination in food samples, which owns great potential for wide-ranging application in harmful compounds assay by simply changing the type of aptamer and its complementary single-stranded DNA.
Collapse
Affiliation(s)
- Rongfang Yan
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Ning Zhang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Weihua Liu
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Xuelian Hu
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Wenxiu Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Yiwei Tang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China.
| | - Shuo Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China; Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Xianghong Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| | - Qinghai Sheng
- College of Food Science and Technology, Hebei Agricultural University, Baoding 071001, China
| |
Collapse
|
6
|
Tetterton-Kellner J, Jensen BC, Nguyen J. Navigating cancer therapy induced cardiotoxicity: From pathophysiology to treatment innovations. Adv Drug Deliv Rev 2024; 211:115361. [PMID: 38901637 PMCID: PMC11534294 DOI: 10.1016/j.addr.2024.115361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/16/2024] [Indexed: 06/22/2024]
Abstract
Every year, more than a million people in the United States undergo chemotherapy or radiation therapy for cancer, as estimated by the CDC. While chemotherapy has been an instrumental tool for treating cancer, it also causes severe adverse effects. The more commonly acknowledged adverse effects include hair loss, fatigue, and nausea, but a more severe and longer lasting side effect is cardiotoxicity. Cardiotoxicity, or heart damage, is a common complication of cancer treatments. It can range from mild to severe, and it can affect some patients temporarily or others permanently, even after they are cured of cancer. Dexrazoxane is the only FDA-approved drug for treating anthracycline induced cardiotoxicity, but it also has drawbacks and adverse effects. There is no other type of chemotherapy induced cardiotoxicity that has an approved treatment option. In this review, we discuss the pathophysiology of chemotherapeutic-induced cardiotoxicity, methods and guidelines of diagnosis, methods of treatment and mitigation, and current drug delivery approaches in therapeutic development.
Collapse
Affiliation(s)
- Jessica Tetterton-Kellner
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Brian C Jensen
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Medicine, Division of Cardiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
7
|
Sabatelle RC, Colson YL, Sachdeva U, Grinstaff MW. Drug Delivery Opportunities in Esophageal Cancer: Current Treatments and Future Prospects. Mol Pharm 2024; 21:3103-3120. [PMID: 38888089 PMCID: PMC11331583 DOI: 10.1021/acs.molpharmaceut.4c00246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
With one of the highest mortality rates of all malignancies, the 5-year survival rate for esophageal cancer is under 20%. Depending on the stage and extent of the disease, the current standard of care treatment paradigm includes chemotherapy or chemoradiotherapy followed by surgical esophagogastrectomy, with consideration for adjuvant immunotherapy for residual disease. This regimen has high morbidity, due to anatomic changes inherent in surgery, the acuity of surgical complications, and off-target effects of systemic chemotherapy and immunotherapy. We begin with a review of current treatments, then discuss new and emerging targets for therapies and advanced drug delivery systems. Recent and ongoing preclinical and early clinical studies are evaluating traditional tumor targets (e.g., human epidermal growth factor receptor 2), as well as promising new targets such as Yes-associated protein 1 or mammalian target of rapamycin to develop new treatments for this disease. Due the function and location of the esophagus, opportunities also exist to pair these treatments with a drug delivery strategy to increase tumor targeting, bioavailability, and intratumor concentrations, with the two most common delivery platforms being stents and nanoparticles. Finally, early results with antibody drug conjugates and chimeric antigenic receptor T cells show promise as upcoming therapies. This review discusses these innovations in therapeutics and drug delivery in the context of their successes and failures, with the goal of identifying those solutions that demonstrate the most promise to shift the paradigm in treating this deadly disease.
Collapse
Affiliation(s)
- Robert C. Sabatelle
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Uma Sachdeva
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Mark W. Grinstaff
- Departments of Biomedical Engineering and Chemistry, Boston University, Boston, MA, 02215, USA
| |
Collapse
|
8
|
Chuang WH, Pislyagin E, Lin LY, Menchinskaya E, Chernikov O, Kozhemyako V, Gorpenchenko T, Manzhulo I, Chaikina E, Agafonova I, Silchenko A, Avilov S, Stonik V, Tzou SC, Aminin D, Wang YM. Holothurian triterpene glycoside cucumarioside A 2-2 induces macrophages activation and polarization in cancer immunotherapy. Cancer Cell Int 2023; 23:292. [PMID: 38001420 PMCID: PMC10668486 DOI: 10.1186/s12935-023-03141-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Despite intensive developments of adoptive T cell and NK cell therapies, the efficacy against solid tumors remains elusive. Our study demonstrates that macrophage-based cell therapy could be a potent therapeutic option against solid tumors. METHODS To this end, we determine the effect of a natural triterpene glycoside, cucumarioside A2-2 (CA2-2), on the polarization of mouse macrophages into the M1 phenotype, and explore the antitumor activity of the polarized macrophage. The polarization of CA2-2-pretreated macrophages was analyzed by flow cytometry and confocal imaging. The anti-cancer activity of CA2-2 macrophages was evaluated against 4T1 breast cancer cells and EAC cells in vitro and syngeneic mouse model in vivo. RESULTS Incubation of murine macrophages with CA2-2 led to polarization into the M1 phenotype, and the CA2-2-pretreated macrophages could selectively target and kill various types of cancer in vitro. Notably, loading near-infrared (NIR) fluorochrome-labeled nanoparticles, MnMEIO-mPEG-CyTE777, into macrophages substantiated that M1 macrophages can target and penetrate tumor tissues in vivo efficiently. CONCLUSION In this study, CA2-2-polarized M1 macrophages significantly attenuated tumor growth and prolonged mice survival in the syngeneic mouse models. Therefore, ex vivo CA2-2 activation of mouse macrophages can serve as a useful model for subsequent antitumor cellular immunotherapy developments.
Collapse
Affiliation(s)
- Wen-Han Chuang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS²B), National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
| | - Evgeny Pislyagin
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Liang-Yu Lin
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
| | - Ekaterina Menchinskaya
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Oleg Chernikov
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Valery Kozhemyako
- Pacific State Medical University, Ostryakova Avenue, Building 2, Vladivostok, 690002, Russia
| | - Tatiana Gorpenchenko
- Federal Scientific Center of East Asia Terrestrial Biodiversity, Far Eastern Branch of the Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Igor Manzhulo
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of the Russian Academy of Science, Palchevskogo str. 17, Vladivostok, 690041, Russia
| | - Elena Chaikina
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Irina Agafonova
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Alexandra Silchenko
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Sergey Avilov
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Valentin Stonik
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia
| | - Shey-Cherng Tzou
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS²B), National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan
| | - Dmitry Aminin
- Far Eastern Branch, G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Russian Academy of Science, 159, Pr. 100 let Vladivostoku, Vladivostok, 690022, Russia.
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, No. 100, Shin-Chuan 1st Road, Sanmin District, Kaohsiung City, 80708, Taiwan.
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan.
- Center for Intelligent Drug Systems and Smart Bio-devices (IDS²B), National Yang Ming Chiao Tung University, Hsinchu, 300, Taiwan.
| |
Collapse
|
9
|
Xiao Q, Zhang Y, Zhao A, Duan Z, Yao J. Application and development of nanomaterials in the diagnosis and treatment of esophageal cancer. Front Bioeng Biotechnol 2023; 11:1268454. [PMID: 38026877 PMCID: PMC10657196 DOI: 10.3389/fbioe.2023.1268454] [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: 07/28/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Esophageal cancer is a malignant tumor with a high incidence worldwide. Currently, there are a lack of effective early diagnosis and treatment methods for esophageal cancer. However, delivery systems based on nanoparticles (NPs) have shown ideal efficacy in real-time imaging and chemotherapy, radiotherapy, gene therapy, and phototherapy for tumors, which has led to their recent widespread design as novel treatment strategies. Compared to traditional drugs, nanomedicine has unique advantages, including strong targeting ability, high bioavailability, and minimal side effects. This article provides an overview of the application of NPs in the diagnosis and treatment of esophageal cancer and provides a reference for future research.
Collapse
Affiliation(s)
| | | | | | | | - Jun Yao
- Henan Key Laboratory of Cancer Epigenetics, Cancer Institute, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
10
|
Anwar S, Khawar MB, Afzal A, Ovais M, Xiang Z. Self-assembled and Zn(II)-coordinated dipeptide nanoparticles with membrane-rupturing action on bacteria. Appl Microbiol Biotechnol 2023; 107:5775-5787. [PMID: 37439833 DOI: 10.1007/s00253-023-12648-4] [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: 11/10/2022] [Revised: 04/07/2023] [Accepted: 06/15/2023] [Indexed: 07/14/2023]
Abstract
Metal ion-coordinated self-assembled short-chain amino acid peptide molecules with multi-photon excitation wavelengths and their photoluminescence properties are advantageous for fluorescence-based diagnostics and treatments of biological diseases based on their extra features of antibacterial agents. We have designed a novel strategy based on tryptophan molecule coordinated with Zn(II) ions in the form of biocompatible spherical nanoparticles of diameter 30-80 nm which have been used for antibacterial treatments against different kinds of pathogenic bacteria (Escherichia coli, Salmonella typhimurium, and Pseudomonas). Preferably, we have used tryptophan-phenylalanine (Trp-Phe), a dipeptide molecule having tryptophan as principal material against E. coli strains as antimicrobial agents for surface rupturing and killing purposes. Furthermore, based on single amino acid, tryptophan, self-assembled and Zn(II)-coordinated dipeptide nanoparticles (Zn-DPNPs) were studied against three types of multi-drug-resistant bacteria as an active antimicrobial agent. These antibacterial efficient nanoparticles may have best alternative of antibiotic drugs for clinical applications. The capability of self-assembled fluorescence behavior of Zn-coordinated dipeptide molecules and higher hydrophobicity against bacterial cell wall will perform as antimicrobial fluorescent agents. KEY POINTS: • Zn(II) and Cu(II) better coordinated into self-assembled NPs. • Fluorescence signals showed interaction of NPs with gram -ve cell wall. • Significant surface-damaging effects were observed in the case of Cu-DPNPs and Zn-DPNPs.
Collapse
Affiliation(s)
- Shahzad Anwar
- National Institutes of Lasers and Optronics College, Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan.
- University of Chinese Academy of Sciences, PR, 100049, Beijing, China.
| | - Muhammad Babar Khawar
- University of Chinese Academy of Sciences, PR, 100049, Beijing, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan
| | - Ali Afzal
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Muhammad Ovais
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology (NCNST), PR, 100190, Beijing, China
| | - Zhang Xiang
- University of Chinese Academy of Sciences, PR, 100049, Beijing, China
| |
Collapse
|
11
|
Deng D, Chang Y, Liu W, Ren M, Xia N, Hao Y. Advancements in Biosensors Based on the Assembles of Small Organic Molecules and Peptides. BIOSENSORS 2023; 13:773. [PMID: 37622859 PMCID: PMC10452798 DOI: 10.3390/bios13080773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023]
Abstract
Over the past few decades, molecular self-assembly has witnessed tremendous progress in a variety of biosensing and biomedical applications. In particular, self-assembled nanostructures of small organic molecules and peptides with intriguing characteristics (e.g., structure tailoring, facile processability, and excellent biocompatibility) have shown outstanding potential in the development of various biosensors. In this review, we introduced the unique properties of self-assembled nanostructures with small organic molecules and peptides for biosensing applications. We first discussed the applications of such nanostructures in electrochemical biosensors as electrode supports for enzymes and cells and as signal labels with a large number of electroactive units for signal amplification. Secondly, the utilization of fluorescent nanomaterials by self-assembled dyes or peptides was introduced. Thereinto, typical examples based on target-responsive aggregation-induced emission and decomposition-induced fluorescent enhancement were discussed. Finally, the applications of self-assembled nanomaterials in the colorimetric assays were summarized. We also briefly addressed the challenges and future prospects of biosensors based on self-assembled nanostructures.
Collapse
Affiliation(s)
- Dehua Deng
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Yong Chang
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Wenjing Liu
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Mingwei Ren
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Ning Xia
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Yuanqiang Hao
- School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan 411201, China
| |
Collapse
|
12
|
Cheng M, Xin Q, Ma S, Ge M, Wang F, Yan X, Jiang B. Advances in the Theranostics of Oesophageal Squamous Carcinoma. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202200251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Indexed: 01/04/2025]
Abstract
AbstractOesophageal squamous carcinoma (ESCC) is one of the most lethal human malignancies, and it is a more aggressive form of oesophageal cancer (EC) that comprises over 90% of all EC cases in China compared with oesophageal adenocarcinoma (EAC). The high mortality of ESCC is attributed to the late‐stage diagnosis, chemoradiotherapy resistance, and lack of appropriate therapeutic targets and corresponding therapeutic formulations. Recently, emerging clinical and translational investigations have involved genome analyses, diagnostic biomarkers, and targeted therapy for ESCC, and these studies provide a new horizon for improving the clinical outcomes of patients with ESCC. Here, the latest research advances in the theranostics of ESCC are reviewed and the unique features of ESCC (including differences from EAC, genomic alterations, and microbe infections), tissue and circulating biomarkers, chemoradiotherapy resistance, clinical targeted therapy for ESCC, identification of novel therapeutic targets, and designation of nanotherapeutic systems for ESCC are particularly focused on. Finally, the perspectives for future clinical and translational theranostic research of ESCC are discussed and the obstacles that must be overcome in ESCC theranostics are described.
Collapse
Affiliation(s)
- Miaomiao Cheng
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Qi Xin
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Saiyu Ma
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Mengyue Ge
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Feng Wang
- Oncology Department The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450000 China
| | - Xiyun Yan
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Protein and Peptide Pharmaceuticals Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| | - Bing Jiang
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
| |
Collapse
|
13
|
Teranishi K. Ready vascular permeability of a near-infrared fluorescent agent ASP5354 for intraoperative ureteral identification enables imaging of carcinoma tissues. Sci Rep 2023; 13:9832. [PMID: 37330535 PMCID: PMC10276870 DOI: 10.1038/s41598-023-37025-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
This study investigates the ability of a near-infrared fluorescence (NIRF) imaging agent, ASP5354, for in vivo fluorescence imaging of esophageal squamous cell carcinoma (ESCC) tissues. The ability of ASP5354 was evaluated using a single dose of ASP5354 or indocyanine green (ICG), which was intravenously administered to a KYSE850 human ESCC xenograft mouse model. Subsequently, in vivo NIRF images of the mouse were obtained using a clinically available camera system. ASP5354-specific NIRF signals were strongly detectable in KYSE850 carcinoma tissues immediately (30 s) following ASP5354 administration compared with normal tissues. Meanwhile, ICG could not distinguish between normal and carcinomatous tissues. To elucidate the associated imaging mechanisms, the vascular permeability of ASP5354 and ICG was investigated in rat back dermis treated with saline or histamine, which enhances vascular permeability, using in vivo NIRF imaging. ASP5354 exhibited higher vascular permeability in histamine-treated skin than in normal skin. KYSE850 carcinoma tissues can be distinguished from normal tissues based on the measurement of ASP5354-specific NIRF signals, and the mechanism that enables imaging relies on the specific and rapid leakage of ASP5354 from the capillaries into the stroma of carcinoma tissues.
Collapse
Affiliation(s)
- Katsunori Teranishi
- Graduate School of Bioresources, Mie University, 1577 Kurimamachiya, Tsu, Mie, 514-8507, Japan.
| |
Collapse
|
14
|
Barman P, Joshi S, Sharma S, Preet S, Sharma S, Saini A. Strategic Approaches to Improvise Peptide Drugs as Next Generation Therapeutics. Int J Pept Res Ther 2023; 29:61. [PMID: 37251528 PMCID: PMC10206374 DOI: 10.1007/s10989-023-10524-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 05/31/2023]
Abstract
In recent years, the occurrence of a wide variety of drug-resistant diseases has led to an increase in interest in alternate therapies. Peptide-based drugs as an alternate therapy hold researchers' attention in various therapeutic fields such as neurology, dermatology, oncology, metabolic diseases, etc. Previously, they had been overlooked by pharmaceutical companies due to certain limitations such as proteolytic degradation, poor membrane permeability, low oral bioavailability, shorter half-life, and poor target specificity. Over the last two decades, these limitations have been countered by introducing various modification strategies such as backbone and side-chain modifications, amino acid substitution, etc. which improve their functionality. This has led to a substantial interest of researchers and pharmaceutical companies, moving the next generation of these therapeutics from fundamental research to the market. Various chemical and computational approaches are aiding the production of more stable and long-lasting peptides guiding the formulation of novel and advanced therapeutic agents. However, there is not a single article that talks about various peptide design approaches i.e., in-silico and in-vitro along with their applications and strategies to improve their efficacy. In this review, we try to bring different aspects of peptide-based therapeutics under one article with a clear focus to cover the missing links in the literature. This review draws emphasis on various in-silico approaches and modification-based peptide design strategies. It also highlights the recent progress made in peptide delivery methods important for their enhanced clinical efficacy. The article would provide a bird's-eye view to researchers aiming to develop peptides with therapeutic applications. Graphical Abstract
Collapse
Affiliation(s)
- Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Shubhi Joshi
- Energy Research Centre, Panjab University, Sector 14, Chandigarh, 160014 India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Simran Preet
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Shweta Sharma
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| |
Collapse
|
15
|
Kong J, Zhao S, Han X, Li W, Zhang J, Wang Y, Shen X, Xia Y, Li Z. Quantitative Ratiometric Biosensors Based on Fluorescent Ferrocene-Modified Histidine Dipeptide Nanoassemblies. Anal Chem 2023; 95:5053-5060. [PMID: 36892972 DOI: 10.1021/acs.analchem.2c05609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Fluorescent proteins (FPs) provide a ratiometric readout for quantitative assessment of the destination of internalized biomolecules. FP-inspired peptide nanostructures that can compete with FPs in their capacity are the most preferred building blocks for the synthesis of fluorescent soft matter. However, realizing a ratiometric emission from a single peptide fluorophore remains exclusive since multicolor emission is a rare property in peptide nanostructures. Here, we describe a bioinspired peptidyl platform for ratiometric intracellular quantitation by employing a single ferrocene-modified histidine dipeptide. The intensiometric ratio of green to blue fluorescence correlates linearly with the concentration of the peptide by three orders of magnitude. The ratiometric fluorescence of the peptide is an assembly-induced emission originating from hydrogen bonds and aromatic interactions. Additionally, modular design enables ferrocene-modified histidine dipeptides to use as a general platform for the construction of intricate peptides that retain the ratiometric fluorescent properties. The ratiometric peptide technique promises flexibility in the design of a wide spectrum of stoichiometric biosensors for quantitatively understanding the trafficking and subcellular fate of biomolecules.
Collapse
Affiliation(s)
- Jia Kong
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Shixuan Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Xue Han
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Wenxin Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Jiaxing Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Yuefei Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, P. R. China
| | - Xihui Shen
- State Key Laboratory of Crop Stress Biology for Arid Areas, Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Yinqiang Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Zhonghong Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| |
Collapse
|
16
|
Kumar V, Ozguney B, Vlachou A, Chen Y, Gazit E, Tamamis P. Peptide Self-Assembled Nanocarriers for Cancer Drug Delivery. J Phys Chem B 2023; 127:1857-1871. [PMID: 36812392 PMCID: PMC10848270 DOI: 10.1021/acs.jpcb.2c06751] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/24/2022] [Indexed: 02/24/2023]
Abstract
The design of novel cancer drug nanocarriers is critical in the framework of cancer therapeutics. Nanomaterials are gaining increased interest as cancer drug delivery systems. Self-assembling peptides constitute an emerging novel class of highly attractive nanomaterials with highly promising applications in drug delivery, as they can be used to facilitate drug release and/or stability while reducing side effects. Here, we provide a perspective on peptide self-assembled nanocarriers for cancer drug delivery and highlight the aspects of metal coordination, structure stabilization, and cyclization, as well as minimalism. We review particular challenges in nanomedicine design criteria and, finally, provide future perspectives on addressing a portion of the challenges via self-assembling peptide systems. We consider that the intrinsic advantages of such systems, along with the increasing progress in computational and experimental approaches for their study and design, could possibly lead to novel classes of single or multicomponent systems incorporating such materials for cancer drug delivery.
Collapse
Affiliation(s)
- Vijay
Bhooshan Kumar
- The
Shmunis School of Biomedicine and Cancer Research, George S. Wise
Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Busra Ozguney
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Anastasia Vlachou
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
| | - Yu Chen
- The
Shmunis School of Biomedicine and Cancer Research, George S. Wise
Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ehud Gazit
- The
Shmunis School of Biomedicine and Cancer Research, George S. Wise
Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Department
of Materials Science and Engineering, Iby and Aladar Fleischman Faculty
of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol
School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Phanourios Tamamis
- Artie
McFerrin Department of Chemical Engineering, Texas A&M University, College Station, Texas 77843-3122, United States
- Department
of Materials Science and Engineering, Texas
A&M University, College
Station, Texas 77843-3003, United States
| |
Collapse
|
17
|
Subhan MA, Parveen F, Filipczak N, Yalamarty SSK, Torchilin VP. Approaches to Improve EPR-Based Drug Delivery for Cancer Therapy and Diagnosis. J Pers Med 2023; 13:jpm13030389. [PMID: 36983571 PMCID: PMC10051487 DOI: 10.3390/jpm13030389] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The innovative development of nanomedicine has promised effective treatment options compared to the standard therapeutics for cancer therapy. However, the efficiency of EPR-targeted nanodrugs is not always pleasing as it is strongly prejudiced by the heterogeneity of the enhanced permeability and retention effect (EPR). Targeting the dynamics of the EPR effect and improvement of the therapeutic effects of nanotherapeutics by using EPR enhancers is a vital approach to developing cancer therapy. Inadequate data on the efficacy of EPR in humans hampers the clinical translation of cancer drugs. Molecular targeting, physical amendment, or physiological renovation of the tumor microenvironment (TME) are crucial approaches for improving the EPR effect. Advanced imaging technologies for the visualization of EPR-induced nanomedicine distribution in tumors, and the use of better animal models, are necessary to enhance the EPR effect. This review discusses strategies to enhance EPR effect-based drug delivery approaches for cancer therapy and imaging technologies for the diagnosis of EPR effects. The effort of studying the EPR effect is beneficial, as some of the advanced nanomedicine-based EPR-enhancing approaches are currently undergoing clinical trials, which may be helpful to improve EPR-induced drug delivery and translation to clinics.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
- Correspondence: (M.A.S.); (V.P.T.)
| | - Farzana Parveen
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Punjab 63100, Pakistan
- Department of Pharmacy Services, DHQ Hospital Jhang 35200, Primary and Secondary Healthcare Department, Government of Punjab, Lahore, Punjab 54000, Pakistan
| | - Nina Filipczak
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | | | - Vladimir P. Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
- Correspondence: (M.A.S.); (V.P.T.)
| |
Collapse
|
18
|
Advances in Self-Assembled Peptides as Drug Carriers. Pharmaceutics 2023; 15:pharmaceutics15020482. [PMID: 36839803 PMCID: PMC9964150 DOI: 10.3390/pharmaceutics15020482] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/19/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
In recent years, self-assembled peptide nanotechnology has attracted a great deal of attention for its ability to form various regular and ordered structures with diverse and practical functions. Self-assembled peptides can exist in different environments and are a kind of medical bio-regenerative material with unique structures. These materials have good biocompatibility and controllability and can form nanoparticles, nanofibers and hydrogels to perform specific morphological functions, which are widely used in biomedical and material science fields. In this paper, the properties of self-assembled peptides, their influencing factors and the nanostructures that they form are reviewed, and the applications of self-assembled peptides as drug carriers are highlighted. Finally, the prospects and challenges for developing self-assembled peptide nanomaterials are briefly discussed.
Collapse
|
19
|
Gao Y, Wang K, Zhang J, Duan X, Sun Q, Men K. Multifunctional nanoparticle for cancer therapy. MedComm (Beijing) 2023; 4:e187. [PMID: 36654533 PMCID: PMC9834710 DOI: 10.1002/mco2.187] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 01/14/2023] Open
Abstract
Cancer is a complex disease associated with a combination of abnormal physiological process and exhibiting dysfunctions in multiple systems. To provide effective treatment and diagnosis for cancer, current treatment strategies simultaneously focus on various tumor targets. Based on the rapid development of nanotechnology, nanocarriers have been shown to exhibit excellent potential for cancer therapy. Compared with nanoparticles with single functions, multifunctional nanoparticles are believed to be more aggressive and potent in the context of tumor targeting. However, the development of multifunctional nanoparticles is not simply an upgraded version of the original function, but involves a sophisticated system with a proper backbone, optimized modification sites, simple preparation method, and efficient function integration. Despite this, many well-designed multifunctional nanoparticles with promising therapeutic potential have emerged recently. Here, to give a detailed understanding and analyzation of the currently developed multifunctional nanoparticles, their platform structures with organic or inorganic backbones were systemically generalized. We emphasized on the functionalization and modification strategies, which provide additional functions to the nanoparticle. We also discussed the application combination strategies that were involved in the development of nanoformulations with functional crosstalk. This review thus provides an overview of the construction strategies and application advances of multifunctional nanoparticles.
Collapse
Affiliation(s)
- Yan Gao
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Kaiyu Wang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Jin Zhang
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Xingmei Duan
- Department of PharmacyPersonalized Drug Therapy Key Laboratory of Sichuan ProvinceSichuan Academy of Medical Sciences & Sichuan Provincial People's HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaChengduSichuan ProvinceChina
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| | - Ke Men
- State Key Laboratory of Biotherapy and Cancer CenterWest China Hospital of Sichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
20
|
Luan X, Kong H, He P, Yang G, Zhu D, Guo L, Wei G. Self-Assembled Peptide-Based Nanodrugs: Molecular Design, Synthesis, Functionalization, and Targeted Tumor Bioimaging and Biotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205787. [PMID: 36440657 DOI: 10.1002/smll.202205787] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/14/2022] [Indexed: 06/16/2023]
Abstract
Functional nanomaterials as nanodrugs based on the self-assembly of inorganics, polymers, and biomolecules have showed wide applications in biomedicine and tissue engineering. Ascribing to the unique biological, chemical, and physical properties of peptide molecules, peptide is used as an excellent precursor material for the synthesis of functional nanodrugs for highly effective cancer therapy. Herein, recent progress on the design, synthesis, functional regulation, and cancer bioimaging and biotherapy of peptide-based nanodrugs is summarized. For this aim, first molecular design and controllable synthesis of peptide nanodrugs with 0D to 3D structures are presented, and then the functional customization strategies for peptide nanodrugs are presented. Then, the applications of peptide-based nanodrugs in bioimaging, chemotherapy, photothermal therapy (PTT), and photodynamic therapy (PDT) are demonstrated and discussed in detail. Furthermore, peptide-based drugs in preclinical, clinical trials, and approved are briefly described. Finally, the challenges and potential solutions are pointed out on addressing the questions of this promising research topic. This comprehensive review can guide the motif design and functional regulation of peptide nanomaterials for facile synthesis of nanodrugs, and further promote their practical applications for diagnostics and therapy of diseases.
Collapse
Affiliation(s)
- Xin Luan
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Hao Kong
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Peng He
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Guozheng Yang
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Danzhu Zhu
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| | - Lei Guo
- Institute of Biomedical Engineering, College of Life Science, Qingdao University, Qingdao, 266071, P. R. China
| | - Gang Wei
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, P. R. China
| |
Collapse
|
21
|
Guan X, Lan T, Wang Y, Cui Y, Duan J, Xu H. CircKRT14 upregulates E2F3 by interacting with miR-1256 to act as an oncogenic factor in esophageal cancer. Hum Exp Toxicol 2023; 42:9603271231155093. [PMID: 36738282 DOI: 10.1177/09603271231155093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND A growing number of studies have focused on the regulatory role of circular RNAs (circRNAs) in a variety of cancers. The purpose of this study was to investigate the effect of circRNA Keratin 14 (circKRT14) on the progression of esophageal cancer (EC). METHODS The levels of circKRT14, miR-1256 and E2F transcription factor 3 (E2F3) were analyzed by real-time quantitative polymerase chain reaction (qRT-PCR) and western blot. The circular structure of circKRT14 was confirmed by RNase R digestion assay. Cell apoptosis, migration and invasion were detected by flow cytometry and transwell assay. The protein levels of related factors were determined by western blot. The relationship between miR-1256 and circKRT14 or E2F3 was verified by dual-luciferase reporter assay. The in vivo function of circKRT14 was studied by xenograft tumor assay. RESULTS CircKRT14 was significantly increased in EC tissues and cells. CircKRT14 silencing inhibited EC cell proliferation, migration, and invasion, but promoted EC cell apoptosis in vitro. CircKRT1 acted as a sponge for miR-1256 in EC, and in-miR-1256 abolished the inhibitory effect of circKRT14 suppression on EC cell progression. E2F3 was a target of miR-1256 and functioned as an oncogene in EC cells. MiR-1256 curbed EC progression by downregulating E2F3. CircKRT14 could affect E2F3 expression by targeting miR-1256. CircKRT14 regulated EC progression in vivo through miR-1256/E2F3 axis. CONCLUSIONS These results uncovered that circKRT14 up-regulated the expression of E2F3 and promoted the malignant development of EC through sponging miR-1256.
Collapse
Affiliation(s)
- Xingzhuo Guan
- Department of Gastroenterology, Affiliated Hospital of Beihua University, Jilin, China
| | - Tingzhu Lan
- Department of Gastroenterology, Affiliated Hospital of Beihua University, Jilin, China
| | - Yuanshi Wang
- Department of Gastroenterology, Affiliated Hospital of Beihua University, Jilin, China
| | - Yan Cui
- Department of Gastroenterology, Affiliated Hospital of Beihua University, Jilin, China
| | - Jinyu Duan
- Department of Gastroenterology, Affiliated Hospital of Beihua University, Jilin, China
| | - Hongjun Xu
- Department of Gastroenterology, 604073The People's Hospital of Suzhou New District, Suzhou, China
| |
Collapse
|
22
|
Sun R, Xiang J, Zhou Q, Piao Y, Tang J, Shao S, Zhou Z, Bae YH, Shen Y. The tumor EPR effect for cancer drug delivery: Current status, limitations, and alternatives. Adv Drug Deliv Rev 2022; 191:114614. [PMID: 36347432 DOI: 10.1016/j.addr.2022.114614] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 11/08/2022]
Abstract
Over the past three decades, the enhanced permeability and retention (EPR) effect has been considered the basis of tumor-targeted drug delivery. Various cancer nanomedicines, including macromolecular drugs, have been designed to utilize this mechanism for preferential extravasation and accumulation in solid tumors. However, such nanomedicines have not yet achieved convincing therapeutic benefits in clinics. Increasing evidence suggests that the EPR effect is over-represented in human tumors, especially in metastatic tumors. This review covers the evolution of the concept, the heterogeneity and limitation of the EPR effect in clinical realities, and prospects for alternative strategies independent of the EPR effect.
Collapse
Affiliation(s)
- Rui Sun
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; Department of Cell Biology, School of Basic Medical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China; ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| | - You Han Bae
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA.
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
23
|
Hou L, Hou Y, Liang Y, Chen B, Zhang X, Wang Y, Zhou K, Zhong T, Long B, Pang W, Wang L, Han X, Li L, Xu C, Gross I, Gaiddon C, Fu W, Yao H, Meng X. Anti-tumor effects of P-LPK-CPT, a peptide-camptothecin conjugate, in colorectal cancer. Commun Biol 2022; 5:1248. [PMID: 36376440 PMCID: PMC9663589 DOI: 10.1038/s42003-022-04191-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
To explore highly selective targeting molecules of colorectal cancer (CRC) is a challenge. We previously identified a twelve-amino acid peptide (LPKTVSSDMSLN, namely P-LPK) by phage display technique which may specifically binds to CRC cells. Here we show that P-LPK selectively bind to a panel of human CRC cell lines and CRC tissues. In vivo, Gallium-68 (68Ga) labeled P-LPK exhibits selective accumulation at tumor sites. Then, we designed a peptide-conjugated drug comprising P-LPK and camptothecin (CPT) (namely P-LPK-CPT), and found P-LPK-CPT significantly inhibits tumor growth with fewer side effects in vitro and in vivo. Furthermore, through co-immunoprecipitation and molecular docking experiment, the glutamine transporter solute carrier 1 family member 5 (SLC1A5) was identified as the possible target of P-LPK. The binding ability of P-LPK and SLC1A5 is verified by surface plasmon resonance and immunofluorescence. Taken together, P-LPK-CPT is highly effective for CRC and deserves further development as a promising anti-tumor therapeutic for CRC, especially SLC1A5-high expression type. A peptide that specifically targets amino acid transporter SLC1A5 in colorectal cancer cells is identified and conjugated with camptothecin to show selective cytotoxicity to colorectal cancer cells in preclinical models.
Collapse
|
24
|
Yan R, Wen Z, Hu X, Wang W, Meng H, Song Y, Wang S, Tang Y. A sensitive sensing system based on fluorescence dipeptide nanoparticles for sulfadimethoxine determination. Food Chem 2022; 405:134963. [DOI: 10.1016/j.foodchem.2022.134963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 11/07/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022]
|
25
|
Uno K, Koike T, Hatta W, Saito M, Tanabe M, Masamune A. Development of Advanced Imaging and Molecular Imaging for Barrett's Neoplasia. Diagnostics (Basel) 2022; 12:2437. [PMID: 36292126 PMCID: PMC9600913 DOI: 10.3390/diagnostics12102437] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/04/2022] [Indexed: 11/17/2022] Open
Abstract
Barrett esophagus (BE) is a precursor to a life-threatening esophageal adenocarcinoma (EAC). Surveillance endoscopy with random biopsies is recommended for early intervention against EAC, but its adherence in the clinical setting is poor. Dysplastic lesions with flat architecture and patchy distribution in BE are hardly detected by high-resolution endoscopy, and the surveillance protocol entails issues of time and labor and suboptimal interobserver agreement for diagnosing dysplasia. Therefore, the development of advanced imaging technologies is necessary for Barrett's surveillance. Recently, non-endoscopic or endoscopic technologies, such as cytosponge, endocytoscopy, confocal laser endomicroscopy, autofluorescence imaging, and optical coherence tomography/volumetric laser endomicroscopy, were developed, but most of them are not clinically available due to the limited view field, expense of the equipment, and significant time for the learning curve. Another strategy is focused on the development of molecular biomarkers, which are also not ready to use. However, a combination of advanced imaging techniques together with specific biomarkers is expected to identify morphological abnormalities and biological disorders at an early stage in the surveillance. Here, we review recent developments in advanced imaging and molecular imaging for Barrett's neoplasia. Further developments in multiple biomarker panels specific for Barrett's HGD/EAC include wide-field imaging systems for targeting 'red flags', a high-resolution imaging system for optical biopsy, and a computer-aided diagnosis system with artificial intelligence, all of which enable a real-time and accurate diagnosis of dysplastic BE in Barrett's surveillance and provide information for precision medicine.
Collapse
Affiliation(s)
- Kaname Uno
- Division of Gastroenterology, Tohoku University Hospital, Sendai 981-8574, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Hou L, Zhong T, Cheng P, Long B, Shi L, Meng X, Yao H. Self-assembled peptide-paclitaxel nanoparticles for enhancing therapeutic efficacy in colorectal cancer. Front Bioeng Biotechnol 2022; 10:938662. [PMID: 36246349 PMCID: PMC9554092 DOI: 10.3389/fbioe.2022.938662] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Chemotherapy is one of the main treatments for colorectal cancer, but systemic toxicity severely limits its clinical use. Packaging hydrophobic chemotherapeutic drugs in targeted nanoparticles greatly improve their efficacy and reduce side effects. We previously identified a novel colorectal cancer specific binding peptide P-LPK (LPKTVSSDMSLN) from phage display peptide library. Here we designed a self-assembled paclitaxel (PTX)-loaded nanoparticle (LPK-PTX NPs). LPK-PTX NPs displayed a superior intracellular internalization and improved tumor cytotoxicity in vitro. Cy5.5-labeled LPK-PTX NPs showed much higher tumor accumulation in colorectal cancer-bearing mice. Furthermore, LPK-PTX NPs exhibit enhanced antitumor activity and decreased systemic toxicity in colorectal cancer patient-derived xenografts (PDX) model. The excellent in vitro and in vivo antitumor efficacy proves the improved targeting drug delivery, suggesting that peptide P-LPK has potential to provide a novel approach for enhanced drug delivery with negligible systemic toxicity.
Collapse
Affiliation(s)
- Lidan Hou
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The Digestive Disease Research and Clinical Transformation Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Diseases, Shanghai, China
| | - Ting Zhong
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The Digestive Disease Research and Clinical Transformation Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Diseases, Shanghai, China
| | - Peng Cheng
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastroenterology, Hainan West Central Hospital, Hainan, China
| | - Bohan Long
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The Digestive Disease Research and Clinical Transformation Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Diseases, Shanghai, China
| | - Leilei Shi
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiangjun Meng
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The Digestive Disease Research and Clinical Transformation Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Diseases, Shanghai, China
| | - Han Yao
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The Digestive Disease Research and Clinical Transformation Center, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Diseases, Shanghai, China
- *Correspondence: Han Yao,
| |
Collapse
|
27
|
Huang H, Kiick KL. Peptide-based assembled nanostructures that can direct cellular responses. Biomed Mater 2022; 17. [DOI: 10.1088/1748-605x/ac92b5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/16/2022] [Indexed: 11/12/2022]
Abstract
Abstract
Natural originated materials have been well-studied over the past several decades owing to their higher biocompatibility compared to the traditional polymers. Peptides, consisting of amino acids, are among the most popular programable building blocks, which is becoming a growing interest in nanobiotechnology. Structures assembled using those biomimetic peptides allow the exploration of chemical sequences beyond those been routinely used in biology. In this Review, we discussed the most recent experimental discoveries on the peptide-based assembled nanostructures and their potential application at the cellular level such as drug delivery. In particular, we explored the fundamental principles of peptide self-assembly and the most recent development in improving their interactions with biological systems. We believe that as the fundamental knowledge of the peptide assemblies evolves, the more sophisticated and versatile nanostructures can be built, with promising biomedical applications.
Collapse
|
28
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
29
|
Zeng Y, Qu X, Nie B, Mu Z, Li C, Li G. An electrochemical biosensor based on electroactive peptide nanoprobes for the sensitive analysis of tumor cells. Biosens Bioelectron 2022; 215:114564. [PMID: 35853325 DOI: 10.1016/j.bios.2022.114564] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022]
Abstract
Peptides possess many appealing and desirable features, which have attracted increasing attention in the field of electrochemical biosensing. However, peptides hardly produce noticeable electronic signals in response to target binding events. In this work, amphipathic peptides FFFGGGGRGDS with both target recognition and self-assembly capabilities are designed to be co-assembled with the electroactive species ferrocenecarboxylic acid (FcCOOH). Furthermore, the resultant electroactive peptide nanoprobes (ePNPs) are applied for sensitive electrochemical analysis of tumor cells. Specifically, tumor cells are captured by the electrode modified with the corresponding DNA aptamers, and ePNPs can then selectively bind to integrin proteins on the cell surface, thereby accompanied by a remarkable increase of electrochemical signal. Taking the assay of MDA-MB-231 cells, the fabricated biosensor can detect cancer cells with a detection limit of 7 cells mL-1. Moreover, the ePNPs can act as a universal probe for the detection of different cell lines. Given the merits of easy synthesis, convenient operation, and favorable analytical performance, the proposed biosensor exhibits great potential in developing peptide-based electrochemical biosensing for clinical applications.
Collapse
Affiliation(s)
- Yujing Zeng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Xinyu Qu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Beibei Nie
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Zheying Mu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Chao Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, PR China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
30
|
Kumar S. Meet the Editorial Board Member. Curr Pharm Biotechnol 2022. [DOI: 10.2174/138920102309220331155203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
Approaches to Improve Macromolecule and Nanoparticle Accumulation in the Tumor Microenvironment by the Enhanced Permeability and Retention Effect. Polymers (Basel) 2022; 14:polym14132601. [PMID: 35808648 PMCID: PMC9268820 DOI: 10.3390/polym14132601] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 12/17/2022] Open
Abstract
Passive targeting is the foremost mechanism by which nanocarriers and drug-bearing macromolecules deliver their payload selectively to solid tumors. An important driver of passive targeting is the enhanced permeability and retention (EPR) effect, which is the cornerstone of most carrier-based tumor-targeted drug delivery efforts. Despite the huge number of publications showcasing successes in preclinical animal models, translation to the clinic has been poor, with only a few nano-based drugs currently being used for the treatment of cancers. Several barriers and factors have been adduced for the low delivery efficiency to solid tumors and poor clinical translation, including the characteristics of the nanocarriers and macromolecules, vascular and physiological barriers, the heterogeneity of tumor blood supply which affects the homogenous distribution of nanocarriers within tumors, and the transport and penetration depth of macromolecules and nanoparticles in the tumor matrix. To address the challenges associated with poor tumor targeting and therapeutic efficacy in humans, the identified barriers that affect the efficiency of the enhanced permeability and retention (EPR) effect for macromolecular therapeutics and nanoparticle delivery systems need to be overcome. In this review, approaches to facilitate improved EPR delivery outcomes and the clinical translation of novel macromolecular therapeutics and nanoparticle drug delivery systems are discussed.
Collapse
|
32
|
Zhao Y, Liu L, Liu S, Wang Y, Li Y, Zhang XD. Electronic and Near-Infrared-II Optical Properties of I-Doped Monolayer MoTe 2: A First-Principles Study. ACS OMEGA 2022; 7:11956-11963. [PMID: 35449971 PMCID: PMC9016853 DOI: 10.1021/acsomega.2c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/15/2022] [Indexed: 06/14/2023]
Abstract
Near-infrared-II (NIR-II, 1000-1700 nm) fluorescence imaging is widely used for in vivo biological imaging. With the unique electronic structures and capability of band-gap engineering, two-dimensional (2D) materials can be potential candidates for NIR-II imaging. Herein, a theoretical investigation of the electronic structure and optical properties of iodine (I)-doped monolayer MoTe2 systems with different doping concentrations is carried out through simulations to explore their NIR optical properties. The results suggest that the emergence of impurity levels due to I doping effectively reduces the bandwidth of I-doped monolayer MoTe2 systems, and the bandwidth decreases with the increase in the I doping concentration. Although the I and Mo atoms possess clear covalent-bonding features according to the charge density difference, impurity levels induced by the strong hybridization between the I 5p and Mo 4d orbitals cross the Fermi level, making the doped systems exhibit metallic behavior. In addition, with the increase in the I doping concentration, the energy required for electron transition from valence bands to impurity levels gradually decreases, which can be linked to the enhancement of the optical absorption in the red-shifted NIR-II region. Meanwhile, with a higher I doping concentration, the emission spectra, which are the product of the absorption spectra and quasi-Fermi distributions for electrons and holes, can be enhanced in the NIR-II window.
Collapse
Affiliation(s)
- Yue Zhao
- Tianjin
Key Laboratory of Brain Science and Neural Engineering, Academy of
Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Ling Liu
- Tianjin
Key Laboratory of Brain Science and Neural Engineering, Academy of
Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Department
of Physics, Shanxi Medical University, Taiyuan 030001, China
| | - Shuangjie Liu
- Tianjin
Key Laboratory of Brain Science and Neural Engineering, Academy of
Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Yang Wang
- Tianjin
Key Laboratory of Brain Science and Neural Engineering, Academy of
Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Yonghui Li
- Department
of Physics and Tianjin Key Laboratory of Low Dimensional Materials
Physics and Preparing Technology, Institute of Advanced Materials
Physics, School of Sciences, Tianjin University, Tianjin 300350, China
| | - Xiao-Dong Zhang
- Tianjin
Key Laboratory of Brain Science and Neural Engineering, Academy of
Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
- Department
of Physics and Tianjin Key Laboratory of Low Dimensional Materials
Physics and Preparing Technology, Institute of Advanced Materials
Physics, School of Sciences, Tianjin University, Tianjin 300350, China
| |
Collapse
|
33
|
Sivagnanam S, Das K, Basak M, Mahata T, Stewart A, Maity B, Das P. Self-assembled dipeptide based fluorescent nanoparticles as a platform for developing cellular imaging probes and targeted drug delivery chaperones. NANOSCALE ADVANCES 2022; 4:1694-1706. [PMID: 36134376 PMCID: PMC9417502 DOI: 10.1039/d1na00885d] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/13/2022] [Indexed: 06/16/2023]
Abstract
Self-assembled peptide-based nanostructures, comprised of naturally occurring amino acids, display excellent biocompatibility, biodegradability, flexible responsiveness, and synthetic feasibility and can be customized for various biomedical applications. However, the lack of inherent optical properties of peptide-based nanoparticles is a limitation on their use as imaging probes or drug delivery vehicles. To overcome this impediment, we generated Boc protected tyrosine-tryptophan dipeptide-based nanoparticles (DPNPs) with structure rigidification by Zn(ii), which shifted the peptide's intrinsic fluorescent properties from the ultraviolet to the visible range. These DPNPs are photostable, biocompatible and have visible fluorescence signals that allow for real-time monitoring of their entry into cells. We further show that two DPNPs (PS1-Zn and PS2-Zn) can encapsulate the chemotherapeutic drug doxorubicin (Dox) and facilitate intracellular drug delivery resulting in cancer cell killing actions comparable to the unencapsulated drug. Finally, we chemically modified our DPNPs with an aptamer directed toward the epithelial cell surface marker EPCAM, which improved Dox delivery to the lung cancer epithelial cell line A549. In contrast, the aptamer conjugated DPNPs failed to deliver Dox into the cardiomyocyte cell line AC16. Theoretically, this strategy could be employed in vivo to specifically deliver Dox to cancer cells while sparing the myocardium, a major source of dose-limiting adverse events in the clinic. Our work represents an important proof-of-concept exercise demonstrating that ultra-short peptide-based fluorescent nanostructures have great promise for the development of new imaging probes and targeted drug delivery vehicles.
Collapse
Affiliation(s)
- Subramaniyam Sivagnanam
- Department of Chemistry, SRM Institute of Science and Technology SRM Nagar, Potheri, Kattankulathur Tamil Nadu 603203 India
| | - Kiran Das
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGI) Campus Raebareli Road Lucknow Uttar Pradesh 226014 India
| | - Madhuri Basak
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGI) Campus Raebareli Road Lucknow Uttar Pradesh 226014 India
| | - Tarun Mahata
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGI) Campus Raebareli Road Lucknow Uttar Pradesh 226014 India
| | - Adele Stewart
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University Jupiter FL 33458 USA
| | - Biswanath Maity
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences (SGPGI) Campus Raebareli Road Lucknow Uttar Pradesh 226014 India
| | - Priyadip Das
- Department of Chemistry, SRM Institute of Science and Technology SRM Nagar, Potheri, Kattankulathur Tamil Nadu 603203 India
| |
Collapse
|
34
|
Jin Y, Yan R, Wang S, Wang X, Zhang X, Tang Y. Dipeptide nanoparticle and aptamer-based hybrid fluorescence platform for enrofloxacin determination. Mikrochim Acta 2022; 189:96. [PMID: 35147788 DOI: 10.1007/s00604-022-05182-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 01/10/2022] [Indexed: 11/25/2022]
Abstract
A novel fluorescence platform was fabricated for enrofloxacin determination by using cDNA-modified dipeptide fluorescence nanoparticles (FDNP-cDNA) and aptamer-modified magnetic Fe3O4 nanoparticles (Fe3O4-Apt). The FDNP were prepared via tryptophan-phenylalanine self-assembling. When magnetic Fe3O4-Apt incubated with standard solution or sample extracts, the target enrofloxacin was selectively captured by the aptamer on the surface of the Fe3O4 nanoparticles. After removing interference by washing with phosphate-buffered saline, the FDNP-cDNA was added, which can bind to the aptamer on the surface of the Fe3O4 nanoparticles not occupied by the analyte. The higher the concentration of the target enrofloxacin in the standard or sample solution is, the less the FDNP-cDNA can be bound with the Fe3O4 nanoparticles, and the more the FDNP-cDNA can be observed in the supernatant. Fluorescence intensity (Ex/Em = 310/380 nm) increased linearly in the enrofloxacin concentration range 0.70 to 10.0 ng/mL with a detection limit of 0.26 ng/mL (S/N = 3). Good recoveries (88.17-99.30%) were obtained in spiked lake water, chicken, and eel samples with relative standard deviation of 2.7-6.2% (n = 3).
Collapse
Affiliation(s)
- Yuting Jin
- College of Food Science and Technology, Hebei Agricultural University, Baoding, 071001, China.,College of Food Science & Project Engineering, Bohai University, Jinzhou, 121013, China
| | - Rongfang Yan
- College of Food Science and Technology, Hebei Agricultural University, Baoding, 071001, China
| | - Shuo Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, 071001, China.,Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
| | - Xianghong Wang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, 071001, China
| | - Xuemei Zhang
- College of Forestry, Hebei Agricultural University, Baoding, 071001, China
| | - Yiwei Tang
- College of Food Science and Technology, Hebei Agricultural University, Baoding, 071001, China.
| |
Collapse
|
35
|
Torrenegra-Rico JD, Arango-Restrepo A, Rubí JM. Enhancing particle transport in deformable micro-channels. J Chem Phys 2022; 156:054118. [DOI: 10.1063/5.0080125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- J. D. Torrenegra-Rico
- Departament de Física de la Matèria Condensada, Facultat de Física, Universitat de Barcelona, Avinguda Diagonal 647, 08028 Barcelona, Spain
| | - A. Arango-Restrepo
- Departament de Física de la Matèria Condensada, Facultat de Física, Universitat de Barcelona, Avinguda Diagonal 647, 08028 Barcelona, Spain
| | - J. M. Rubí
- Departament de Física de la Matèria Condensada, Facultat de Física, Universitat de Barcelona, Avinguda Diagonal 647, 08028 Barcelona, Spain
| |
Collapse
|
36
|
Rehman MU, Khan A, Imtiyaz Z, Ali S, Makeen HA, Rashid S, Arafah A. Current Nano-therapeutic Approaches Ameliorating Inflammation in Cancer Progression. Semin Cancer Biol 2022; 86:886-908. [DOI: 10.1016/j.semcancer.2022.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 01/22/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
|
37
|
Marcazzan S, Braz Carvalho MJ, Konrad M, Strangmann J, Tenditnaya A, Baumeister T, Schmid RM, Wester HJ, Ntziachristos V, Gorpas D, Wang TC, Schottelius M, Quante M. CXCR4 peptide-based fluorescence endoscopy in a mouse model of Barrett's esophagus. EJNMMI Res 2022; 12:2. [PMID: 35006394 PMCID: PMC8748556 DOI: 10.1186/s13550-021-00875-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/24/2021] [Indexed: 12/14/2022] Open
Abstract
Background Near-infrared (NIR) fluorescence imaging has been emerging as a promising strategy to overcome the high number of early esophageal adenocarcinomas missed by white light endoscopy and random biopsy collection. We performed a preclinical assessment of fluorescence imaging and endoscopy using a novel CXCR4-targeted fluorescent peptide ligand in the L2-IL1B mouse model of Barrett’s esophagus. Methods Six L2-IL1B mice with advanced stage of disease (12–16 months old) were injected with the CXCR4-targeted, Sulfo-Cy5-labeled peptide (MK007), and ex vivo wide-field imaging of the whole stomach was performed 4 h after injection. Before ex vivo imaging, fluorescence endoscopy was performed in three L2-IL1B mice (12–14 months old) by a novel imaging system with two L2-IL1B mice used as negative controls. Results Ex vivo imaging and endoscopy in L2-IL1B mice showed that the CXCR4-targeted MK007 accumulated mostly in the dysplastic lesions with a mean target-to-background ratio > 2. The detection of the Sulfo-Cy5 signal in dysplastic lesions and its co-localization with CXCR4 stained cells by confocal microscopy further confirmed the imaging results. Conclusions This preliminary preclinical study shows that CXCR4-targeted fluorescence endoscopy using MK007 can detect dysplastic lesions in a mouse model of Barrett’s esophagus. Further investigations are needed to assess its use in the clinical setting. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-021-00875-7.
Collapse
Affiliation(s)
- Sabrina Marcazzan
- II Medizinische Klinik, Klinikum rechts der isar, Technische Universität München, Munich, Germany.,Chair of Biological Imaging, School of Medicine, Technische Universität München, Munich, Germany.,Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany.,Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University of Innsbruck, Peter-Mayr-Straße 4b, 6020, Innsbruck, Austria
| | - Marcos J Braz Carvalho
- II Medizinische Klinik, Klinikum rechts der isar, Technische Universität München, Munich, Germany
| | - Matthias Konrad
- Institut für Pharmazeutische Radiochemie, Technische Universität München, Munich, Germany
| | - Julia Strangmann
- II Medizinische Klinik, Klinikum rechts der isar, Technische Universität München, Munich, Germany.,Innere Medizin II, Universitätsklinik Freiburg, Universität Freiburg, Freiburg im Breisgau, Germany
| | - Anna Tenditnaya
- Chair of Biological Imaging, School of Medicine, Technische Universität München, Munich, Germany.,Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
| | - Theresa Baumeister
- II Medizinische Klinik, Klinikum rechts der isar, Technische Universität München, Munich, Germany
| | - Roland M Schmid
- II Medizinische Klinik, Klinikum rechts der isar, Technische Universität München, Munich, Germany
| | - Hans-Jürgen Wester
- Institut für Pharmazeutische Radiochemie, Technische Universität München, Munich, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging, School of Medicine, Technische Universität München, Munich, Germany.,Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
| | - Dimitris Gorpas
- Chair of Biological Imaging, School of Medicine, Technische Universität München, Munich, Germany.,Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
| | - Timothy C Wang
- Division of Digestive and Liver Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Margret Schottelius
- Institut für Pharmazeutische Radiochemie, Technische Universität München, Munich, Germany.,Translational Radiopharmaceutical Sciences, Departments of Nuclear Medicine and Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Michael Quante
- II Medizinische Klinik, Klinikum rechts der isar, Technische Universität München, Munich, Germany. .,Innere Medizin II, Universitätsklinik Freiburg, Universität Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
38
|
Rizvi SFA, Mu S, Zhao C, Zhang H. Fabrication of self-assembled peptide nanoparticles for in vitro assessment of cell apoptosis pathway and in vivo therapeutic efficacy. Mikrochim Acta 2022; 189:53. [PMID: 34999971 DOI: 10.1007/s00604-021-05148-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/07/2021] [Indexed: 12/14/2022]
Abstract
Near-infrared fluorescent (NIRF) dye-coupled self-assembled RGD-linked proapoptotic peptide nanoparticles have been synthesized with spherical shape and size ~ 30-40 nm diameters. The peptide sequence was coupled with cyanine 5.5 probe as NIRF-dye to introduce optical imaging properties and pH-dependent method was used to design Cy5.5 coupled self-assembled peptide nanoparticles (f-SAPNs). This nanoprobe has the ability to target αvβ3-integrin receptor overexpressed on cancer cell's surface with improved internalization capabilities into the mitochondria. The in situ study showed that this peptide sequence has potential to disrupt the mitochondrial membrane efficiently, activating the Caspase-3 enzyme, and ultimately induces cell apoptosis. It has been observed from in vitro study that the degree of apoptosis for f-SAPNs was increased from 25.6% to 96.3%, while decreased degree of necrosis from 51.7% to 0.2% compared with its parent peptide analog (Cy5.5-c[RGDKLAK]; f-CP) occurs. Further investigations revealed that these f-SAPNs showed high uptake in U87MG glioblastoma cells in comparison with PC-3 prostate cancer cells. Moreover, in vivo therapeutic studies represented the prominent decrease in the size of tumor tissue treated with f-CP and f-SAPNs (201 ± 13 mm3 and 104 ± 6 mm3, respectively) compared with untreated tumor tissues (366 ± 18 mm3). These outcomes highlighted the specificity, and efficacy of f-SAPNs toward αvβ3-integrin expressing tumor tissue in vivo and suggested that these novel designed f-SAPNs may serve as a potential theranostic drug for brain tumor glioblastoma multiforme. The pH-sensitive method gives NIRF dye-coupled self-assembled peptide nanoparticle (f-SAPNs), enables the tunable synthesis of spherical nanoparticles with high stability towards proteolysis, improved biocompatibility, and promising therapeutic efficacy.
Collapse
Affiliation(s)
- Syed Faheem Askari Rizvi
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Shuai Mu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Chunyan Zhao
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Haixia Zhang
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
| |
Collapse
|
39
|
Sun L. Meet the Editorial Board Member. Curr Pharm Biotechnol 2022. [DOI: 10.2174/138920102301211124143140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Leming Sun
- School of Life Sciences, Northwestern Polytechnical University, Xi’an-710072,China
| |
Collapse
|
40
|
The development and progress of nanomedicine for esophageal cancer diagnosis and treatment. Semin Cancer Biol 2022; 86:873-885. [DOI: 10.1016/j.semcancer.2022.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/22/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
|
41
|
Luo X, Liu J. Ultrasmall Luminescent Metal Nanoparticles: Surface Engineering Strategies for Biological Targeting and Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103971. [PMID: 34796699 PMCID: PMC8787435 DOI: 10.1002/advs.202103971] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/27/2021] [Indexed: 05/07/2023]
Abstract
In the past decade, ultrasmall luminescent metal nanoparticles (ULMNPs, d < 3 nm) have achieved rapid progress in addressing many challenges in the healthcare field because of their excellent physicochemical properties and biological behaviors. With the sharp shrinking size of large plasmonic metal nanoparticles (PMNPs), the contributions from the surface characteristics increase significantly, which brings both opportunities and challenges in the application-driven surface engineering of ULMNPs toward advanced biological applications. Here, the systematic advancements in the biological applications of ULMNPs from bioimaging to theranostics are summarized with emphasis on the versatile surface engineering strategies in the regulation of biological targeting and imaging performance. The efforts in the surface functionalization strategies of ULMNPs for enhanced disease targeting abilities are first discussed. Thereafter, self-assembly strategies of ULMNPs for fabricating multifunctional nanostructures for multimodal imaging and nanomedicine are discussed. Further, surface engineering strategies of ratiometric ULMNPs to enhance the imaging stability to address the imaging challenges in complicated bioenvironments are summarized. Finally, the phototoxicity of ULMNPs and future perspectives are also reviewed, which are expected to provide a fundamental understanding of the physicochemical properties and biological behaviors of ULMNPs to accelerate their future clinical applications in healthcare.
Collapse
Affiliation(s)
- Xiaoxi Luo
- Key Laboratory of Functional Molecular Engineering of Guangdong ProvinceSchool of Chemistry and Chemical EngineeringSouth China University of TechnologyGuangzhou510640China
| | - Jinbin Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong ProvinceSchool of Chemistry and Chemical EngineeringSouth China University of TechnologyGuangzhou510640China
| |
Collapse
|
42
|
Fang HY, Stangl S, Marcazzan S, Carvalho MJB, Baumeister T, Anand A, Strangmann J, Huspenina JS, Wang TC, Schmid RM, Feith M, Friess H, Ntziachristos V, Multhoff G, Gorpas D, Quante M. Targeted Hsp70 fluorescence molecular endoscopy detects dysplasia in Barrett's esophagus. Eur J Nucl Med Mol Imaging 2022; 49:2049-2063. [PMID: 34882260 PMCID: PMC9016004 DOI: 10.1007/s00259-021-05582-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 10/03/2021] [Indexed: 01/21/2023]
Abstract
PURPOSE The incidence of esophageal adenocarcinoma (EAC) has been increasing for decades without significant improvements in treatment. Barrett's esophagus (BE) is best established risk factor for EAC, but current surveillance with random biopsies cannot predict progression to cancer in most BE patients due to the low sensitivity and specificity of high-definition white light endoscopy. METHODS Here, we evaluated the membrane-bound highly specific Hsp70-specific contrast agent Tumor-Penetrating Peptide (Hsp70-TPP) in guided fluorescence molecular endoscopy biopsy. RESULTS Hsp70 was significantly overexpressed as determined by IHC in dysplasia and EAC compared with non-dysplastic BE in patient samples (n = 12) and in high-grade dysplastic lesions in a transgenic (L2-IL1b) mouse model of BE. In time-lapse microscopy, Hsp70-TPP was rapidly taken up and internalized by human BE dysplastic patient-derived organoids. Flexible fluorescence endoscopy of the BE mouse model allowed a specific detection of Hsp70-TPP-Cy5.5 that corresponded closely with the degree of dysplasia but not BE. Ex vivo application of Hsp70-TPP-Cy5.5 to freshly resected whole human EAC specimens revealed a high (> 4) tumor-to-background ratio and a specific detection of previously undetected tumor infiltrations. CONCLUSION In summary, these findings suggest that Hsp70-targeted imaging using fluorescently labeled TPP peptide may improve tumor surveillance in BE patients.
Collapse
Affiliation(s)
- Hsin-Yu Fang
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Stefan Stangl
- Department of Radiation Oncology and Central Institute for Translational Cancer Research, (TranslaTUM), Technische Universität München, Munich, Germany
| | - Sabrina Marcazzan
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany ,Chair of Biological Imaging, School of Medicine, Technische Universität München, Munich, Germany; Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
| | - Marcos J. Braz Carvalho
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Theresa Baumeister
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Akanksha Anand
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Julia Strangmann
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany ,Innere Medizin II, Universitätsklinik Freiburg, Universität Freiburg, Freiburg im Breisgau, Germany
| | | | - Timothy C. Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY USA
| | - Roland M. Schmid
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Marcus Feith
- Chirurgische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Helmut Friess
- Chirurgische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Vasilis Ntziachristos
- Chair of Biological Imaging, School of Medicine, Technische Universität München, Munich, Germany; Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology and Central Institute for Translational Cancer Research, (TranslaTUM), Technische Universität München, Munich, Germany
| | - Dimitris Gorpas
- Chair of Biological Imaging, School of Medicine, Technische Universität München, Munich, Germany; Helmholtz Zentrum München, Institute of Biological and Medical Imaging, Neuherberg, Germany
| | - Michael Quante
- II Medizinische Klinik, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany ,Innere Medizin II, Universitätsklinik Freiburg, Universität Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
43
|
Didamson OC, Abrahamse H. Targeted Photodynamic Diagnosis and Therapy for Esophageal Cancer: Potential Role of Functionalized Nanomedicine. Pharmaceutics 2021; 13:1943. [PMID: 34834358 PMCID: PMC8625244 DOI: 10.3390/pharmaceutics13111943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/26/2021] [Accepted: 10/31/2021] [Indexed: 12/11/2022] Open
Abstract
Esophageal cancer is often diagnosed at the late stage when cancer has already spread and is characterized by a poor prognosis. Therefore, early diagnosis is vital for a better and efficient treatment outcome. Upper endoscopy with biopsy is the standard diagnostic tool for esophageal cancer but is challenging to diagnose at its premalignant stage, while conventional treatments such as surgery, chemotherapy, and irradiation therapy, are challenging to eliminate the tumor. Photodynamic diagnosis (PDD) and therapy (PDT) modalities that employ photosensitizers (PSs) are emerging diagnostic and therapeutic strategies for esophageal cancer. However, some flaws associated with the classic PSs have limited their clinical applications. Functionalized nanomedicine has emerged as a potential drug delivery system to enhance PS drug biodistribution and cellular internalization. The conjugation of PSs with functionalized nanomedicine enables increased localization within esophageal cancer cells due to improved solubility and stability in blood circulation. This review highlights PS drugs used for PDD and PDT for esophageal cancer. In addition, it focuses on the various functionalized nanomedicine explored for esophageal cancer and their role in targeted PDD and PDT for diagnosis and treatment.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, Johannesburg 2028, South Africa;
| |
Collapse
|
44
|
Recent advances in active targeting of nanomaterials for anticancer drug delivery. Adv Colloid Interface Sci 2021; 296:102509. [PMID: 34455211 DOI: 10.1016/j.cis.2021.102509] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/24/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
One of the challenges in cancer chemotherapy is the low target to non-target ratio of therapeutic agents which incur severe adverse effect on the healthy tissues. In this regard, nanomaterials have tremendous potential for impacting cancer therapy by altering the toxicity profile of the drug. Some of the striking advantages provided by the nanocarriers mediated targeted drug delivery are relatively high build-up of drug concentration at the tumor site, improved drug content in the formulation and enhanced colloidal stability. Further, nanocarriers with tumor-specific moieties can be targeted to the cancer cell through cell surface receptors, tumor antigens and tumor vasculatures with high affinity and accuracy. Moreover, it overcomes the bottleneck of aimless drug biodistribution, undesired toxicity and heavy dosage of administration. This review discusses the recent developments in active targeting of nanomaterials for anticancer drug delivery through cancer cell surface targeting, organelle specific targeting and tumor microenvironment targeting strategies. Special emphasis has been given towards cancer cell surface and organelle specific targeting as delivery of anticancer drugs through these routes have made paradigm change in cancer management. Further, the current challenges and future prospects of nanocarriers mediated active drug targeting are also demonstrated.
Collapse
|
45
|
Liu F, Wu Q, Han W, Laster K, Hu Y, Ma F, Chen H, Tian X, Qiao Y, Liu H, Kim DJ, Dong Z, Liu K. Targeting integrin αvβ3 with indomethacin inhibits patient-derived xenograft tumour growth and recurrence in oesophageal squamous cell carcinoma. Clin Transl Med 2021; 11:e548. [PMID: 34709754 PMCID: PMC8552524 DOI: 10.1002/ctm2.548] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE A high risk of post-operative recurrence contributes to the poor prognosis and low survival rate of oesophageal squamous cell carcinoma (ESCC) patients. Increasing experimental evidence suggests that integrin adhesion receptors, in particular integrin αv (ITGAV), are important for cancer cell survival, proliferation and migration. Therefore, targeting ITGAV may be a rational approach for preventing ESCC recurrence. MATERIALS AND METHODS Protein levels of ITGAV were determined in human ESCC tumour tissues using immunohistochemistry. MTT, propidium iodide staining, and annexin V staining were utilized to investigate cell viability, cell cycle progression, and induction of apoptosis, respectively. Computational docking was performed with the Schrödinger Suite software to visualize the interaction between indomethacin and ITGAV. Cell-derived xenograft mouse models, patient-derived xenograft (PDX) mouse models, and a humanized mouse model were employed for in vivo studies. RESULTS ITGAV was upregulated in human ESCC tumour tissues and increased ITGAV protein levels were associated with poor prognosis. ITGAV silencing or knockout suppressed ESCC cell growth and metastatic potential. Interestingly, we identified that indomethacin can bind to ITGAV and enhance synovial apoptosis inhibitor 1 (SYVN1)-mediated degradation of ITGAV. Integrin β3, one of the β subunits of ITGAV, was also decreased at the protein level in the indomethacin treatment group. Importantly, indomethacin treatment suppressed ESCC tumour growth and prevented recurrence in a PDX mouse model. Moreover, indomethacin inhibited the activation of cytokine TGFβ, reduced SMAD2/3 phosphorylation, and increased anti-tumour immune responses in a humanized mouse model. CONCLUSION ITGAV is a promising therapeutic target for ESCC. Indomethacin can attenuate ESCC growth through binding to ITGAV, promoting SYVN1-mediated ubiquitination of ITGAV, and potentiating cytotoxic CD8+ T cell responses.
Collapse
Affiliation(s)
- Fangfang Liu
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Qiong Wu
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Wei Han
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Kyle Laster
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Yamei Hu
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Fayang Ma
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Hanyong Chen
- Hormel InstituteUniversity of MinnesotaAustinMinnesotaUSA
| | - Xueli Tian
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Yan Qiao
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
| | - Hui Liu
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Dong Joon Kim
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
| | - Zigang Dong
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
- State Key Laboratory of Esophageal Cancer Prevention and TreatmentZhengzhouChina
- Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouChina
- Cancer Chemoprevention International Collaboration LaboratoryZhengzhouChina
| | - Kangdong Liu
- Department of PathophysiologySchool of Basic Medical SciencesChina‐US (Henan) Hormel Cancer InstituteAMS, College of MedicineZhengzhou UniversityZhengzhouChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouChina
- State Key Laboratory of Esophageal Cancer Prevention and TreatmentZhengzhouChina
- Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouChina
- Cancer Chemoprevention International Collaboration LaboratoryZhengzhouChina
| |
Collapse
|
46
|
Liu Z, Xie F, Xie J, Chen J, Li Y, Lin Q, Luo F, Yan J. New-generation photosensitizer-anchored gold nanorods for a single near-infrared light-triggered targeted photodynamic-photothermal therapy. Drug Deliv 2021; 28:1769-1784. [PMID: 34470548 PMCID: PMC8425697 DOI: 10.1080/10717544.2021.1960923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Traditional combined photodynamic and photothermal therapy (PDT/PTT) was limited in clinical treatment of cancer due to the exceptionally low drug delivery efficiency to tumor sites and the activation by laser excitation with different wavelengths. We have accidentally discovered that our synthesized chlorin e6-C-15-ethyl ester (HB, a new type of photosensitizer) be activated by a laser with an excitation wavelength of 660 nm. Herein, we utilized Au nanorods (AuNRs) as 660 nm-activated PTT carriers to be successively surface-functionalized with HB and tumor-targeting peptide cyclic RGD (cRGD) to develop HB-AuNRs@cRGD for single NIR laser-induced targeted PDT/PTT. The HB-AuNRs@cRGD could be preferentially accumulated within tumor sites and rapidly internalized by cancer cells. Thereby, the HB-AuNRs@cRGD could exhibit amplified therapeutic effects by producing both significant reactive oxygen species (ROS) and hyperthermia simultaneously under the guidance of fluorescence imaging. The tumor inhibition rate on ECA109 esophageal cancer model was approximately 77.04%, and the negligible systematic toxicity was observed. This study proposed that HB-AuNRs@cRGD might be a promising strategy for single NIR laser-induced and imaging-guided targeted bimodal phototherapy.
Collapse
Affiliation(s)
- Zongjunlin Liu
- School of Medicine, Anti-Cancer Center, Xiamen University, Xiamen, China
| | - Fang Xie
- Department of Radiation Oncology, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Jun Xie
- School of Medicine, Anti-Cancer Center, Xiamen University, Xiamen, China
| | - Jianhao Chen
- Department of Radiation Oncology, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yang Li
- Xiamen Institute of Rare Earth Materials, Institute of Haixi, Chinese Academy of Sciences, Xiamen, China
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Fanghong Luo
- School of Medicine, Anti-Cancer Center, Xiamen University, Xiamen, China
| | - Jianghua Yan
- School of Medicine, Anti-Cancer Center, Xiamen University, Xiamen, China
| |
Collapse
|
47
|
Synthesis, Characterization and Evaluation of Peptide Nanostructures for Biomedical Applications. Molecules 2021; 26:molecules26154587. [PMID: 34361740 PMCID: PMC8348434 DOI: 10.3390/molecules26154587] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/04/2021] [Accepted: 07/17/2021] [Indexed: 12/19/2022] Open
Abstract
There is a challenging need for the development of new alternative nanostructures that can allow the coupling and/or encapsulation of therapeutic/diagnostic molecules while reducing their toxicity and improving their circulation and in-vivo targeting. Among the new materials using natural building blocks, peptides have attracted significant interest because of their simple structure, relative chemical and physical stability, diversity of sequences and forms, their easy functionalization with (bio)molecules and the possibility of synthesizing them in large quantities. A number of them have the ability to self-assemble into nanotubes, -spheres, -vesicles or -rods under mild conditions, which opens up new applications in biology and nanomedicine due to their intrinsic biocompatibility and biodegradability as well as their surface chemical reactivity via amino- and carboxyl groups. In order to obtain nanostructures suitable for biomedical applications, the structure, size, shape and surface chemistry of these nanoplatforms must be optimized. These properties depend directly on the nature and sequence of the amino acids that constitute them. It is therefore essential to control the order in which the amino acids are introduced during the synthesis of short peptide chains and to evaluate their in-vitro and in-vivo physico-chemical properties before testing them for biomedical applications. This review therefore focuses on the synthesis, functionalization and characterization of peptide sequences that can self-assemble to form nanostructures. The synthesis in batch or with new continuous flow and microflow techniques will be described and compared in terms of amino acids sequence, purification processes, functionalization or encapsulation of targeting ligands, imaging probes as well as therapeutic molecules. Their chemical and biological characterization will be presented to evaluate their purity, toxicity, biocompatibility and biodistribution, and some therapeutic properties in vitro and in vivo. Finally, their main applications in the biomedical field will be presented so as to highlight their importance and advantages over classical nanostructures.
Collapse
|
48
|
Yang PP, Li YJ, Cao Y, Zhang L, Wang JQ, Lai Z, Zhang K, Shorty D, Xiao W, Cao H, Wang L, Wang H, Liu R, Lam KS. Rapid discovery of self-assembling peptides with one-bead one-compound peptide library. Nat Commun 2021; 12:4494. [PMID: 34301935 PMCID: PMC8302598 DOI: 10.1038/s41467-021-24597-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/28/2021] [Indexed: 12/02/2022] Open
Abstract
Self-assembling peptides have shown tremendous potential in the fields of material sciences, nanoscience, and medicine. Because of the vast combinatorial space of even short peptides, identification of self-assembling sequences remains a challenge. Herein, we develop an experimental method to rapidly screen a huge array of peptide sequences for self-assembling property, using the one-bead one-compound (OBOC) combinatorial library method. In this approach, peptides on beads are N-terminally capped with nitro-1,2,3-benzoxadiazole, a hydrophobicity-sensitive fluorescence molecule. Beads displaying self-assembling peptides would fluoresce under aqueous environment. Using this approach, we identify eight pentapeptides, all of which are able to self-assemble into nanoparticles or nanofibers. Some of them are able to interact with and are taken up efficiently by HeLa cells. Intracellular distribution varied among these non-toxic peptidic nanoparticles. This simple screening strategy has enabled rapid identification of self-assembling peptides suitable for the development of nanostructures for various biomedical and material applications. Self-assembling peptides have a range of potential applications but developing self-assembling sequences can be challenging. Here, the authors report on a one-bead one-compound combinatorial library where fluorescence is used to detect the potential for self-assembly and identified candidates are evaluated.
Collapse
Affiliation(s)
- Pei-Pei Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, China
| | - Yi-Jing Li
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, China.,Department of Materials Physics and Chemistry, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yan Cao
- Institute for Advanced Study, Shenzhen University, Guangdong, China
| | - Lu Zhang
- Department of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Jia-Qi Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, China
| | - Ziwei Lai
- Institute for Advanced Study, Shenzhen University, Guangdong, China
| | - Kuo Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, China.,Department of Materials Physics and Chemistry, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Diedra Shorty
- Department of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Wenwu Xiao
- Department of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Hui Cao
- Department of Materials Physics and Chemistry, School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, China.
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA.
| | - Kit S Lam
- Department of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA. .,Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, CA, USA.
| |
Collapse
|
49
|
Lin W, Yang Y, Lei Y, An F, Sun L, Qin Y, Zhang L. Self-Assembly of an Antitumor Dipeptide Induced Near-Infrared Fluorescence and Improved Stability for Theranostic Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:32799-32809. [PMID: 34227796 DOI: 10.1021/acsami.1c07983] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
It has been found that the self-assembly of nonfluorescent peptides can generate fluorescent peptide nanoparticles (f-PNPs) to perform multiple functions, including drug delivery and imaging and tracking therapeutic agents. Both pharmacologically inactive peptides and tumor-targeting peptides have been explored to construct biocompatible f-PNPs; however, the application of this technology in delivering antitumor peptides has never been reported. Herein, the self-assembly of an antitumor dipeptide, carnosine, into fluorescent carnosine nanoparticles (f-Car NPs) in the presence of zinc ions is demonstrated. The generated f-Car NPs exhibit fluorescence in the visible and near-infrared (NIR) ranges for fluorescence tracing in vitro and in vivo. On the other hand, the f-Car NPs minimize the contact between the dipeptide and the serum, which overcomes the dipeptide instability resulted from inefficient antitumor activity. In addition, the preparation of f-Car NPs does not introduce extra carrier materials, so the f-Car NPs exhibit biocompatibility to normal fibroblast cells in vitro and negligible toxicity against major organs in vivo. This study provides a new peptide drug delivery strategy with NIR fluorescence tracing ability.
Collapse
Affiliation(s)
- Weifeng Lin
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yingchun Yang
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yang Lei
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an 710072, China
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, China
| | - Leming Sun
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an 710072, China
| | - Yong Qin
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an 710072, China
| | - Lianbing Zhang
- School of Life Sciences, Key Laboratory of Space Bioscience & Biotechnology, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
50
|
Liu M, Fang X, Yang Y, Wang C. Peptide-Enabled Targeted Delivery Systems for Therapeutic Applications. Front Bioeng Biotechnol 2021; 9:701504. [PMID: 34277592 PMCID: PMC8281044 DOI: 10.3389/fbioe.2021.701504] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Receptor-targeting peptides have been extensively pursued for improving binding specificity and effective accumulation of drugs at the site of interest, and have remained challenging for extensive research efforts relating to chemotherapy in cancer treatments. By chemically linking a ligand of interest to drug-loaded nanocarriers, active targeting systems could be constructed. Peptide-functionalized nanostructures have been extensively pursued for biomedical applications, including drug delivery, biological imaging, liquid biopsy, and targeted therapies, and widely recognized as candidates of novel therapeutics due to their high specificity, well biocompatibility, and easy availability. We will endeavor to review a variety of strategies that have been demonstrated for improving receptor-specificity of the drug-loaded nanoscale structures using peptide ligands targeting tumor-related receptors. The effort could illustrate that the synergism of nano-sized structures with receptor-targeting peptides could lead to enrichment of biofunctions of nanostructures.
Collapse
Affiliation(s)
- Mingpeng Liu
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Department of Chemistry, Tsinghua University, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaocui Fang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yanlian Yang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chen Wang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|