1
|
Okafor M, Schmitt D, Ory S, Gasman S, Hureau C, Faller P, Vitale N. The Different Cellular Entry Routes for Drug Delivery Using Cell Penetrating Peptides. Biol Cell 2025; 117:e70012. [PMID: 40490965 DOI: 10.1111/boc.70012] [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: 01/31/2025] [Revised: 04/18/2025] [Accepted: 04/23/2025] [Indexed: 06/11/2025]
Abstract
The cell plasma membrane acts as a semi-permeable barrier essential for cellular protection and function, posing a challenge for therapeutic molecule delivery. Conventional techniques for crossing this barrier, including biophysical and biochemical methods, often exhibit limitations such as cytotoxicity and the risk of genomic integration when viral vectors are involved. In contrast, cell-penetrating peptides (CPPs) offer a promising non-invasive means to deliver a broad range of molecular cargoes, including proteins, nucleic acids and small molecules, into cells. CPPs, typically 5 to 30 amino acids long and rich in basic or non-polar residues, interact favourably with different cell membranes. These peptides have evolved since the discovery of the HIV-1 TAT peptide in the 1980s, expanding into various CPP families with diverse therapeutic applications. CPPs can form covalent or non-covalent complexes with their cargo, influencing their stability and efficacy. Based on their sequence properties and interactions, CPPs can be amphipathic or non-amphipathic, with distinct mechanisms of membrane penetration, such as direct penetration and endocytosis. While their uptake mechanisms are complex and not fully elucidated, ongoing optimization aims to enhance CPP specificity and efficacy. CPPs have demonstrated potential in drug delivery, gene therapy, cancer treatment and vaccine development, addressing key safety and efficiency concerns associated with viral vectors. This review explores the classification, mechanisms of action and therapeutic potential. It focuses on the intracellular vesicular trafficking of CPPs, highlighting their role as transformative tools in advancing cellular therapies and medical treatments.
Collapse
Affiliation(s)
- Michael Okafor
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
- Institut de Chimie-UMR7177, Université de Strasbourg/CNRS, Strasbourg, France
| | - David Schmitt
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
- Laboratoire de Chimie de Coordination-CNRS UPR8241, Université de Toulouse, Toulouse, France
| | - Stéphane Ory
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
| | - Stéphane Gasman
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
| | - Christelle Hureau
- Laboratoire de Chimie de Coordination-CNRS UPR8241, Université de Toulouse, Toulouse, France
| | - Peter Faller
- Institut de Chimie-UMR7177, Université de Strasbourg/CNRS, Strasbourg, France
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives-CNRS UPR3212, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
2
|
Wang D, Yin F, Li Z, Zhang Y, Shi C. Current progress and remaining challenges of peptide-drug conjugates (PDCs): next generation of antibody-drug conjugates (ADCs)? J Nanobiotechnology 2025; 23:305. [PMID: 40259322 PMCID: PMC12013038 DOI: 10.1186/s12951-025-03277-2] [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: 12/26/2024] [Accepted: 02/24/2025] [Indexed: 04/23/2025] Open
Abstract
Drug conjugates have emerged as a promising alternative delivery system designed to deliver an ultra-toxic payload directly to the target cancer cells, maximizing therapeutic efficacy while minimizing toxicity. Among these, antibody-drug conjugates (ADCs) have garnered significant attention from both academia and industry due to their great potential for cancer therapy. However, peptide-drug conjugates (PDCs) offer several advantages over ADCs, including more accessible industrial synthesis, versatile functionalization, high tissue penetration, and rapid clearance with low immunotoxicity. These factors position PDCs as up-and-coming drug candidates for future cancer therapy. Despite their potential, PDCs face challenges such as poor pharmacokinetic properties and low bioactivity, which hinder their clinical development. How to design PDCs to meet clinical needs is a big challenge and urgent to resolve. In this review, we first carefully analyzed the general consideration of successful PDC design learning from ADCs. Then, we summarised the basic functions of each component of a PDC construct, comprising of peptides, linkers and payloads. The peptides in PDCs were categorized into three types: tumor targeting peptides, cell penetrating peptide and self-assembling peptide. We then analyzed the potential of these peptides for drug delivery, such as overcoming drug resistance, controlling drug release and improving therapeutic efficacy with reduced non-specific toxicity. To better understand the potential druggability of PDCs, we discussed the pharmacokinetics of PDCs and also briefly introduced the current PDCs in clinical trials. Lastly, we discussed the future perspectives for the successful development of an oncology PDC. This review aimed to provide useful information for better construction of PDCs in future clinical applications.
Collapse
Affiliation(s)
- Dongyuan Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Feng Yin
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
- Shenzhen Bay Laboratory, Pingshan Translational Medicine Center, Shenzhen, 518118, China
| | - Zigang Li
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
- Shenzhen Bay Laboratory, Pingshan Translational Medicine Center, Shenzhen, 518118, China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China.
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China.
| |
Collapse
|
3
|
Wu Y, Chen Z, Chen F, Su J, Han J, Liu S. Using Regular Porous Membrane-Based Blood-Brain Barrier Model to Screen Brain-Targeted Drugs with Nanochannel Electrochemistry. Anal Chem 2025; 97:7968-7977. [PMID: 40183921 DOI: 10.1021/acs.analchem.5c00365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Constructing in vitro blood-brain barrier (BBB) model provides an innovative approach for studying the pathophysiology of the brain and screening drugs. Although commercial Transwell was the simplest and most widely utilized in vitro model, reasonably mimicking essential characteristics of human BBB and dynamic monitoring BBB function remain a challenge. Herein, inspired by the highly permeable extracellular matrix membrane in human BBB, a novel in vitro BBB model was established by combining functionalized anodic alumina oxide (AAO) membrane with nanochannel electrochemistry (ANE-BBB). Benefiting from the topographical nanostructures and modified cell-adhesive peptide on the AAO surface, a tight endothelial barrier was formed, which can be directly visualized by phase-contrast microscope, and the barrier function can be real time monitored by nanochannel electrochemistry. More importantly, according to the current signal induced by the diffusion of redox species through the nanochannels toward the underlying electrode surface, dynamic evaluation of BBB-crossing behavior and precise screening of brain-targeted nanodrugs can be achieved. The constructed ANE-BBB overcomes the shortcomings of invisible cell culture, low permeability, and inaccurate real-time monitoring of screening drugs in traditional Transwell and provides a reliable tool for the design of nanodrugs to the central nervous system.
Collapse
Affiliation(s)
- Yafeng Wu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Zixuan Chen
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Fengxiang Chen
- State Key Laboratory of New Textile Materials and Advanced Processing, Wuhan Textile University, Wuhan 430200, China
| | - Juan Su
- Targeted MRI Contrast Agents Laboratory of Jiangsu Province, Nanjing Polytechnic Institute, Nanjing 210048, China
| | - Jianyu Han
- School of Energy and Environment, Southeast University, Nanjing 210096, China
| | - Songqin Liu
- Jiangsu Engineering Laboratory of Smart Carbon-Rich Materials and Device, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
4
|
Zhou S, Zhang M, Wang J, Chen X, Xu Z, Yan Y, Li Y. Nanofibers in Glioma Therapy: Advances, Applications, and Overcoming Challenges. Int J Nanomedicine 2025; 20:4677-4703. [PMID: 40255668 PMCID: PMC12008729 DOI: 10.2147/ijn.s510363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/31/2025] [Indexed: 04/22/2025] Open
Abstract
Despite relentless effort to study glioma treatment, the prognosis for glioma patients remains poor. The main obstacles include the high rate of recurrence and the difficulty of passing the blood-brain barrier (BBB) for therapeutic drugs. Nanomaterials owing to their special physicochemical properties have been used in a wide range of fields thus far. The nanodrug delivery system (NDDS) with the ability of crossing the BBB, targeting glioma site, maintaining drug stability and controlling drug release, has significantly enhanced the anti-tumor therapeutic effect, improving the prognosis of glioma patients. Aligned nanofibers (NFs) are ideal materials to establish in vitro models of glioma microenvironment (GME), enabling the exploration of the mechanism of glioma cell migration and invasion to discover novel therapeutic targets. Moreover, NFs are now widely used in glioma applications such as radiotherapy, phototherapy, thermotherapy and immunotherapy. Despite the absolute dominance of NFs in anti-glioma applications, there are still some problems such as the further optimization of NDDS, and the impact of interactions between nanofibers and the protein corona (PC) on glioma therapy. This paper will shed light on the latest glioma applications of NFs in drug delivery systems and mimicking the tumor microenvironment (TME), and discuss how to further optimize the NDDS and eliminate or utilize the nanomedicine-PC interactions.
Collapse
Affiliation(s)
- Shangjun Zhou
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, People’s Republic of China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Mingcheng Zhang
- Center of Endoscopy, The Second Affiliated Hospital of Shandong First Medical University Tai’an, Shandong, People’s Republic of China
| | - Jiayu Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xi Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Yong Li
- Department of Pediatric Surgery, Hunan Children’s Hospital, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
5
|
Lu Y, Li G, Zhang Y, Ge Y, Hao B, Yin Y, Zhao Y, Wang Y. Engineered Assemblies from Constitutionally Isomeric Peptides Modulate Antimicrobial Activity. Biomacromolecules 2025; 26:2614-2624. [PMID: 40152418 DOI: 10.1021/acs.biomac.5c00071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Antimicrobial peptides (AMPs) are a class of peptides consisting of cationic amino acid residues and a hydrophobic segment, which have been used as an alternative to antibiotics in treating multidrug-resistant bacteria. However, the relationship among the molecular design, assembled structures, and resultant efficacy remains elusive. Herein, we report a class of constitutionally isomeric AMPs assembled into filaments with similar dimensions. Spectroscopic characterizations demonstrated that subtle changes in the position of amino acids led to dramatic variations in molecular packing and surface charges, which were verified by molecular dynamics simulations. In vitro antibacterial assays showed that all AMPs exerted antibacterial activity against Gram-positive methicillin-resistant Staphylococcus aureus (MRSA), but the efficacy was dependent on the molecular design. Given the good biocompatibility to eukaryotic cells, these AMPs could be potentially used as antibacterial agents. We believe that this finding provides an avenue to tune the bioactivity of AMPs by rational molecular design.
Collapse
Affiliation(s)
- Yujia Lu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guanyi Li
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yanwen Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuxuan Ge
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bin Hao
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Yin
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yaxue Zhao
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yin Wang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, Engineering Research Center of Cell & Therapeutic Antibody, National Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
6
|
Cao M, Wang R, Xu X, Hou X, Wang W, Zhang X, Ma C, Zhang Y, Shi D, Yang J, Ma H. Engineering of peptide assemblies for adaptable protein delivery to achieve efficient intracellular biocatalysis. J Colloid Interface Sci 2025; 683:457-467. [PMID: 39693883 DOI: 10.1016/j.jcis.2024.12.097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
Efficient intracellular delivery of native proteins remains a big challenge, which greatly hinders the development of protein therapy. Here, we report a generalizable peptide vector that can encapsulate and deliver various proteins to achieve efficient intracellular biocatalysis. The peptide was rationally designed to be cationic amphiphilic peptide that consist of four functional fragments, that is, a hydrophobic domain to promote molecular assembly, an enzyme-cleavable fragment to introduce stimuli-responsibility, several cationic arginine (Arg) residues to enhance cell interaction and transmembrane efficiency, and the cystine (Cys) residues with redox sensitivity to adjust the stability of the peptide/protein complexes as needed. The peptide can co-assemble with proteins to form stable complexes in aqueous solution under mild condition. The complexes enter cell mainly through caveolae- and lipid raft-mediated endocytosis, giving a delivery efficiency of up to ∼97.2 %. They can then achieve efficient lysosomal escape and disassociation to release native proteins inside cells in response to intracellular stimuli. More strikingly, the delivered protein's bioactivity can be well maintained and the two model proteins of β-galactosidase (β-Gal) and horseradish peroxidase (HRP) both showed excellent intracellular biocatalytic activity. The study develops a versatile and adjustable peptide carrier platform for protein delivery and highlights impactful structure-function relationships, providing a new chemical guide for the design and optimization of functional protein nanocarriers.
Collapse
Affiliation(s)
- Meiwen Cao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China.
| | - Rui Wang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Xiaomin Xu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Xinyue Hou
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Wentao Wang
- Department of Radiochemistry, China Institute of Atomic Energy, Beijing 102413, China.
| | - Xiaoming Zhang
- School of Science, Optoelectronics Research Center, Minzu University of China, Beijing 100081, China
| | - Chen Ma
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Yuxuan Zhang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Daikui Shi
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Jianing Yang
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| | - Hongchao Ma
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East), 66 Changjiang West Road, Qingdao 266580, China
| |
Collapse
|
7
|
Das TN, Ramesh A, Ghosh A, Moyra S, Maji TK, Ghosh G. Peptide-based nanomaterials and their diverse applications. NANOSCALE HORIZONS 2025; 10:279-313. [PMID: 39629637 DOI: 10.1039/d4nh00371c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The supramolecular self-assembly of peptides offers a promising avenue for both materials science and biological applications. Peptides have garnered significant attention in molecular self-assembly, forming diverse nanostructures with α-helix, β-sheet, and random coil conformations. These self-assembly processes are primarily driven by the amphiphilic nature of peptides and stabilized by non-covalent interactions, leading to complex nanoarchitectures responsive to environmental stimuli. While extensively studied in biomedical applications, including drug delivery and tissue engineering, their potential applications in the fields of piezoresponsive materials, conducting materials, catalysis and energy harvesting remain underexplored. This review comprehensively elucidates the diverse material characteristics and applications of self-assembled peptides. We discuss the multi-stimuli-responsiveness of peptide self-assemblies and their roles as energy harvesters, catalysts, liquid crystalline materials, glass materials and contributors to electrical conductivity. Additionally, we address the challenges and present future perspectives associated with peptide nanomaterials. This review aims to provide insights into the versatile applications of peptide self-assemblies while concisely summarizing their well-established biomedical roles that have previously been extensively reviewed by various research groups, including our group.
Collapse
Affiliation(s)
- Tarak Nath Das
- Molecular Materials Laboratory, New Chemistry Unit (NCU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India.
| | - Aparna Ramesh
- Centre for Nano and Soft Matter Sciences (CeNS), Shivanapura, Dasanapura Hobli, Bengaluru, 562162, India.
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Arghya Ghosh
- Molecular Materials Laboratory, New Chemistry Unit (NCU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India.
| | - Sourav Moyra
- Centre for Nano and Soft Matter Sciences (CeNS), Shivanapura, Dasanapura Hobli, Bengaluru, 562162, India.
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Tapas Kumar Maji
- Molecular Materials Laboratory, New Chemistry Unit (NCU), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India.
- Molecular Materials Laboratory, Chemistry and Physics of Materials Unit (CPMU), International Centre for Materials Science (ICMS), School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur, Bangalore 560064, India
| | - Goutam Ghosh
- Centre for Nano and Soft Matter Sciences (CeNS), Shivanapura, Dasanapura Hobli, Bengaluru, 562162, India.
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
8
|
Jiang S, Zu C, Wang B, Zhong Y. Enhancing DNA Vaccine Delivery Through Stearyl-Modified Cell-Penetrating Peptides: Improved Antigen Expression and Immune Response In Vitro and In Vivo. Vaccines (Basel) 2025; 13:94. [PMID: 39852873 PMCID: PMC11768954 DOI: 10.3390/vaccines13010094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Inefficient cellular uptake is a significant limitation to the efficacy of DNA vaccines. In this study, we introduce S-Cr9T, a stearyl-modified cell-penetrating peptide (CPP) designed to enhance DNA vaccine delivery by forming stable complexes with plasmid DNA, thereby protecting it from degradation and promoting efficient intracellular uptake. METHODS AND RESULTS In vitro studies showed that S-Cr9T significantly improved plasmid stability and transfection efficiency, with optimal performance at an N/P ratio of 0.25. High-content imaging revealed that the S-Cr9T-plasmid complex stably adhered to the cell membrane, leading to enhanced plasmid uptake and transfection. In vivo, S-Cr9T significantly increased antigen expression and triggered a robust immune response, including a threefold increase in IFN-γ secretion and several hundred-fold increases in antibody levels compared to control groups. CONCLUSIONS These findings underscore the potential of S-Cr9T to enhance DNA vaccine efficacy, offering a promising platform for advanced gene therapy and vaccination strategies.
Collapse
Affiliation(s)
- Sheng Jiang
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; (S.J.); (C.Z.)
| | - Cheng Zu
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; (S.J.); (C.Z.)
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Bin Wang
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; (S.J.); (C.Z.)
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yiwei Zhong
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China; (S.J.); (C.Z.)
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
9
|
Bojarska J, Wolf WM. Short Peptides as Powerful Arsenal for Smart Fighting Cancer. Cancers (Basel) 2024; 16:3254. [PMID: 39409876 PMCID: PMC11476321 DOI: 10.3390/cancers16193254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Short peptides have been coming around as a strong weapon in the fight against cancer on all fronts-in immuno-, chemo-, and radiotherapy, and also in combinatorial approaches. Moreover, short peptides have relevance in cancer imaging or 3D culture. Thanks to the natural 'smart' nature of short peptides, their unique structural features, as well as recent progress in biotechnological and bioinformatics development, short peptides are playing an enormous role in evolving cutting-edge strategies. Self-assembling short peptides may create excellent structures to stimulate cytotoxic immune responses, which is essential for cancer immunotherapy. Short peptides can help establish versatile strategies with high biosafety and effectiveness. Supramolecular short peptide-based cancer vaccines entered clinical trials. Peptide assemblies can be platforms for the delivery of antigens, adjuvants, immune cells, and/or drugs. Short peptides have been unappreciated, especially in the vaccine aspect. Meanwhile, they still hide the undiscovered unlimited potential. Here, we provide a timely update on this highly active and fast-evolving field.
Collapse
Affiliation(s)
- Joanna Bojarska
- Chemistry Department, Institute of Inorganic and Ecological Chemistry, Łódź University of Technology, S. Żeromskiego Str. 116, 90-924 Łódź, Poland;
| | | |
Collapse
|
10
|
Sis MJ, Liu D, Allen I, Webber MJ. Iterative Design Reveals Molecular Domain Relationships in Supramolecular Peptide-Drug Conjugates. Biomacromolecules 2024; 25:4482-4491. [PMID: 38870408 DOI: 10.1021/acs.biomac.4c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Supramolecular peptide-drug conjugates (sPDCs) are prepared by covalent attachment of a drug moiety to a peptide motif programmed for one-dimensional self-assembly, with subsequent physical entanglement of these fibrillar structures enabling formation of nanofibrous hydrogels. This class of prodrug materials presents an attractive platform for mass-efficient and site-specific delivery of therapeutic agents using a discrete, single-component molecular design. However, a continued challenge in sPDC development is elucidating relationships between supramolecular interactions in their drug and peptide domains and the resultant impacts of these domains on assembly outcomes and material properties. Inclusion of a saturated alkyl segment alongside the prodrug in the hydrophobic domain of sPDCs could relieve some of the necessity for ordered, prodrug-produg interactions. Accordingly, nine sPDCs are prepared here to iterate the design variables of amino acid sequence and hydrophobic prodrug-alkyl block design. All molecules spontaneously formed hydrogels under physiological conditions, indicating a less hindered thermodynamic path to self-assembly relative to previous prodrug-only designs. However, material studies on the supramolecular arrangement, formation, and mechanical properties of the resultant sPDC hydrogels as well as their drug release profiles showed complex relationships between the hydrophobic and peptide domains in the formation and function of the resulting assemblies. Together, these results indicate that sPDC material properties are intrinsically linked to holistic molecular design with coupled contributions from their prodrug and peptide domains in directing properties of the emergent materials.
Collapse
Affiliation(s)
- Matthew J Sis
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Dongping Liu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Isabella Allen
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana 46556, United States
| |
Collapse
|
11
|
Liu J, Zhang J, Zhang Y, Wei W, Zhan M, Zhang Z, Liu B, Hu X, He W. A mitochondria-targeting heptamethine cyanine-chlorambucil formulated polymeric nanoparticle to potentiate native tumor chemotherapeutic efficacy. Biomater Sci 2024; 12:2614-2625. [PMID: 38591255 DOI: 10.1039/d4bm00003j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2024]
Abstract
Chlorambucil (Cbl) is a DNA alkylating drug in the nitrogen mustard family, but the clinical applications of nitrogen mustard antitumor drugs are frequently limited by their poor aqueous solubility, poor cellular uptake, lack of targeting, and severe side effects. Additionally, mitochondria are the energy factories for cells, and tumor cells are more susceptible to mitochondrial dysfunction than some healthy cells, thus making mitochondria an important target for tumor therapy. As a proof-of-concept, direct delivery of Cbl to tumor cells' mitochondria will probably bring about new opportunities for the nitrogen mustard family. Furthermore, IR775 chloride is a small-molecule lipophilic cationic heptamethine cyanine dye with potential advantages of mitochondria targeting, near-infrared (NIR) fluorescence imaging, and preferential internalization towards tumor cells. Here, an amphiphilic drug conjugate was facilely prepared by covalently coupling chlorambucil with IR775 chloride and further self-assembly to form a carrier-free self-delivery theranostic system, in which the two components are both functional units aimed at theranostic improvement. The theranostic IR775-Cbl potentiated typical "1 + 1 > 2" tumor inhibition through specific accumulation in mitochondria, which triggered a remarkable decrease in mitochondrial membrane potential and ATP generation. In vivo biodistribution and kinetic monitoring were achieved by real-time NIR fluorescence imaging to observe its transport inside a living body. Current facile mitochondria-targeting modification with clinically applied drugs was promising for endowing traditional drugs with targeting, imaging, and improved potency in disease theranostics.
Collapse
Affiliation(s)
- Jing Liu
- College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Jie Zhang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China.
| | - Yongteng Zhang
- Key Laboratory of Precision and Intelligent Chemistry, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, and School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026 Anhui, China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, 215123 Suzhou, China
| | - Wei Wei
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China.
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China.
| | - Zhiren Zhang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China.
| | - Bing Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, 519000, Guangdong, China.
| | - Xianglong Hu
- Key Laboratory of Precision and Intelligent Chemistry, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, and School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026 Anhui, China.
- Suzhou Institute for Advanced Research, University of Science and Technology of China, 215123 Suzhou, China
| | - Weiling He
- Department of Gastrointestinal Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361000, China.
| |
Collapse
|
12
|
Dowaidar M. Uptake pathways of cell-penetrating peptides in the context of drug delivery, gene therapy, and vaccine development. Cell Signal 2024; 117:111116. [PMID: 38408550 DOI: 10.1016/j.cellsig.2024.111116] [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: 01/03/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
Cell-penetrating peptides have been extensively utilized for the purpose of facilitating the intracellular delivery of cargo that is impermeable to the cell membrane. The researchers have exhibited proficient delivery capabilities for oligonucleotides, thereby establishing cell-penetrating peptides as a potent instrument in the field of gene therapy. Furthermore, they have demonstrated a high level of efficiency in delivering several additional payloads. Cell penetrating peptides (CPPs) possess the capability to efficiently transport therapeutic molecules to specific cells, hence offering potential remedies for many illnesses. Hence, their utilization is imperative for the improvement of therapeutic vaccines. In contemporary studies, a plethora of cell-penetrating peptides have been unveiled, each characterized by its own distinct structural attributes and associated mechanisms. Although it is widely acknowledged that there are multiple pathways through which particles might be internalized, a comprehensive understanding of the specific mechanisms by which these particles enter cells has to be fully elucidated. The absorption of cell-penetrating peptides can occur through either direct translocation or endocytosis. However, it is worth noting that categories of cell-penetrating peptides are not commonly linked to specific entrance mechanisms. Furthermore, research has demonstrated that cell-penetrating peptides (CPPs) possess the capacity to enhance antigen uptake by cells and facilitate the traversal of various biological barriers. The primary objective of this work is to examine the mechanisms by which cell-penetrating peptides are internalized by cells and their significance in facilitating the administration of drugs, particularly in the context of gene therapy and vaccine development. The current study investigates the immunostimulatory properties of numerous vaccine components administered using different cell-penetrating peptides (CPPs). This study encompassed a comprehensive discussion on various topics, including the uptake pathways and mechanisms of cell-penetrating peptides (CPPs), the utilization of CPPs as innovative vectors for gene therapy, the role of CPPs in vaccine development, and the potential of CPPs for antigen delivery in the context of vaccine development.
Collapse
Affiliation(s)
- Moataz Dowaidar
- Bioengineering Department, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Interdisciplinary Research Center for Hydrogen Technologies and Carbon Management, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia; Biosystems and Machines Research Center, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran 31261, Saudi Arabia.
| |
Collapse
|
13
|
Gad HA, Diab AM, Elsaied BE, Tayel AA. Biopolymer-based formulations for curcumin delivery toward cancer management. CURCUMIN-BASED NANOMEDICINES AS CANCER THERAPEUTICS 2024:309-338. [DOI: 10.1016/b978-0-443-15412-6.00009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
14
|
Mo Y, Yang Y, Zeng J, Ma W, Guan Y, Guo J, Wu X, Liu D, Feng L, Jia X, Yang B. Enhancing the Biopharmacological Characteristics of Asperosaponin VI: Unveiling Dynamic Self-Assembly Phase Transitions in the Gastrointestinal Environment. Int J Nanomedicine 2023; 18:7335-7358. [PMID: 38084126 PMCID: PMC10710790 DOI: 10.2147/ijn.s436372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
PURPOSE Asperosaponin VI (ASP VI) as an active ingredient of Dipsacus asperoides, which has a wide range of biological and pharmacological activity. However, its development and application are restricted due to the poor gastrointestinal permeability and oral bioavailability. This investigation aims to reveal the influence of the self-assembled structure by the interaction between ASP VI and endogenous components NaTC and/or DOPC in the gastrointestinal environment on its biopharmaceutical properties, and novelty elucidated the molecular mechanism for the formation of self-assembled nanomicelles. METHODS This change in phase state in gastrointestinal fluids is characterized by dynamic light scattering (DLS) and transmission electron microscope (TEM). UPLC-Q-TOF-MS was used to analyze the composition of phase components and the exposure of nanomicelles in vivo. Molecular dynamics simulation (MDS) was applied to preliminarily elucidate the self-assembly mechanism of ASP VI in the gastrointestinal environment. Furthermore, theS8 promoting absorption mechanism of nanomicelles were investigated through in vivo pharmacokinetic experiments, parallel artificial membrane permeability assay (PAMPA), quadruple single-pass intestinal perfusion in rats, and Caco-2 cell monolayer model. RESULTS We demonstrated that the ASP VI could spontaneously form dynamic self-assembled structures with sodium taurocholate (NaTC) and dipalmitoyl phosphatidylcholine (DOPC) during gastrointestinal solubilization, which promoted the gastrointestinal absorption and permeability of ASP VI and increased its exposure in vivo, thus improving the biopharmacological characteristics of ASP VI. Moreover, ASP VI-NaTC-DOPC-self-assembled nanostructures (ASP VI-NaTC-DOPC-SAN) manifested higher cellular uptake in Caco-2 cells as evidenced by flow cytometry and confocal microscopy, and this study also preliminarily revealed the mechanism of self-assembly formation of ASP VI with endogenous components NaTC and DOPC driven by electrostatic and hydrogen bonding interactions. CONCLUSION This study provides evidence that the dynamic self-assembled phase transition may play a key role in improving the biopharmacological characteristics of insoluble or low permeability active ingredients during the gastrointestinal dissolution of Chinese medicines.
Collapse
Affiliation(s)
- Yulin Mo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Yanjun Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Jingqi Zeng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Weikun Ma
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Yuxin Guan
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Jingxi Guo
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Xiaochun Wu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Dingkun Liu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Liang Feng
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Xiaobin Jia
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| | - Bing Yang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, People’s Republic of China
| |
Collapse
|
15
|
Zhang T, Luo X, Xu K, Zhong W. Peptide-containing nanoformulations: Skin barrier penetration and activity contribution. Adv Drug Deliv Rev 2023; 203:115139. [PMID: 37951358 DOI: 10.1016/j.addr.2023.115139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/21/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Transdermal drug delivery presents a less invasive pathway, circumventing the need to pass through the gastrointestinal tract and liver, thereby reducing drug breakdown, initial metabolism, and gastrointestinal discomfort. Nevertheless, the unique composition and dense structure of the stratum corneum present a significant barrier to transdermal delivery. This article presents an overview of the current developments in peptides and nanotechnology to address this challenge. Initially, we sum up peptide-containing nanoformulations for transdermal drug delivery, examining them through the lenses of both inorganic and organic materials. Particular emphasis is placed on the diverse roles that peptides play within these nanoformulations, including conferring functionality upon nanocarriers and enhancing the biological efficacy of drugs. Subsequently, we summarize innovative strategies for enhancing skin penetration, categorizing them into passive and active approaches. Lastly, we discuss the therapeutic potential of peptide-containing nanoformulations in addressing a range of diseases, drawing insights from the biological activities and functions of peptides. Furthermore, the challenges hindering clinical translation are also discussed, providing valuable insights for future advancements in transdermal drug delivery.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xuan Luo
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Keming Xu
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China.
| | - Wenying Zhong
- Department of Chemistry, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Biomedical Functional Materials, China Pharmaceutical University, Nanjing 210009, China; Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
16
|
Sánchez-López E, Gómara MJ, Haro I. Atorvastatin-loaded peptide amphiphiles against corneal neovascularization. Nanomedicine (Lond) 2023; 18:1095-1108. [PMID: 37610088 DOI: 10.2217/nnm-2023-0133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023] Open
Abstract
Background: Corneal neovascularization is a sight-threatening disease. It can be treated using antiangiogenic and anti-inflammatory compounds. Therefore, atorvastatin (ATV) constitutes a suitable candidate to be administered topically. To attain suitable efficacy, ATV can be encapsulated into custom-developed nanocarriers such as peptide amphiphiles. Methods: Three peptide amphiphiles bearing one, two or four C16-alkyl groups (mC16-Tat47-57, dC16-Tat47-57 and qC16-Tat47-57) were synthesized, characterized and loaded with ATV. Drug release and ocular tolerance were assessed as well as anti-inflammatory and antiangiogenic properties. Results: ATV-qC16-Tat47-57 showed higher encapsulation efficiency than mC16-Tat47-57 and dC16-Tat47-57 and more defined nanostructures. ATV-qC16-Tat47-57 showed ATV prolonged release with suitable ocular tolerance. Moreover, ATV-qC16-Tat47-57 was antiangiogenic and prevented ocular inflammation. Conclusion: ATV-qC16-Tat47-57 constitutes a promising topical medication against corneal neovascularization.
Collapse
Affiliation(s)
- Elena Sánchez-López
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain
- Institute of Nanoscience & Nanotechnology (IN2UB), University of Barcelona, 08028, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28031, Madrid, Spain
- Unit of Synthesis & Biomedical applications of Peptides, IQAC-CSIC, 08034, Barcelona, Spain
| | - Maria José Gómara
- Unit of Synthesis & Biomedical applications of Peptides, IQAC-CSIC, 08034, Barcelona, Spain
| | - Isabel Haro
- Unit of Synthesis & Biomedical applications of Peptides, IQAC-CSIC, 08034, Barcelona, Spain
| |
Collapse
|
17
|
Xu Q, Li X, Yang J, Zhang Y, Deng X, Li G, Yuan Q. Naphthyl-Poly(S-((2-carboxyethyl)thio)-l-cysteine) Peptide Amphiphiles with Different Degrees of Polymerization: Synthesis, Self-Assembly, pH/Reduction-Triggered Drug Release, and Cytotoxicity. Mol Pharm 2023; 20:1256-1268. [PMID: 36648435 DOI: 10.1021/acs.molpharmaceut.2c00925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Four peptide amphiphiles (PA1-4) with different degrees of polymerization (DP = 40, 15, 10, and 6) were synthesized by Fuchs-Farthing and ring-opening polymerization followed by post-polymerization modification, as fully characterized by 1H NMR, FT-IR, gel permeation chromatography, and circular dichroism (CD) spectroscopy. It was found that PAs could self-assemble to form regular spherical micelles in low-concentration (about 1 mg/mL) aqueous solution, which had different contents of secondary structures and mainly adopted random coil conformations. The water solubility of PAs increases with the increase of DP, the polypeptide chain stretches randomly in water, the β-sheets decrease, and the random coil conformations dominate. When the pH of PA solution decreases or increases, intramolecular hydrogen bonds break, and molecular chains stretch, leading to a decrease of α-helix, turn conformations, and an increase of β-sheets. Meanwhile, the particle size of micelles increases. At around 0.4 mg/mL, the hemolysis ability of PA2 is negligible at pH 7.4 and 6.5 and about 33% at pH 5.5. Cisplatin (CDDP) was linked to micelles by coordination bonds to explore their potential as drug carriers, exhibiting controlled pH and reduction in dual drug release effects. MTT assay showed that the HeLa cell viability was 78% when cultured in the 13.5 μg/mL PA2 blank micelles for 2 days, while the cell viability was 60% in the CDDP-loaded micelles. Furthermore, a high concentration of PA2 (about 100 mg/mL) could self-assemble into a fibrous hydrogel at pH 5.5, which self-healed 2 h after incision and self-degraded 71% within 14 days. The CDDP-loaded fiber hydrogel exhibited a sustained release effect similar to the CDDP-loaded micelles. The cytotoxicity of CDDP-loaded fibers at 48 h was detected to be the same as that of the same amount of CDDP, and the cell viability was 7%. Therefore, we provide a new strategy for the synthesis of amphiphilic peptides with potential applications in nano-drug carriers and cancer therapy.
Collapse
Affiliation(s)
- Qinming Xu
- School of Materials and Energy, National Center for International Research on Photoelectric and Energy Materials, Yunnan University, Kunming650091, PR China
| | - Xing Li
- School of Materials and Energy, National Center for International Research on Photoelectric and Energy Materials, Yunnan University, Kunming650091, PR China
| | - Jingang Yang
- School of Materials and Energy, National Center for International Research on Photoelectric and Energy Materials, Yunnan University, Kunming650091, PR China
| | - Yan Zhang
- School of Chemical Science and Engineering, Yunnan University, Kunming650091, PR China
| | - Xiaocui Deng
- School of Materials and Energy, National Center for International Research on Photoelectric and Energy Materials, Yunnan University, Kunming650091, PR China
| | - Gang Li
- School of Materials and Energy, National Center for International Research on Photoelectric and Energy Materials, Yunnan University, Kunming650091, PR China
| | - Qingmei Yuan
- School of Materials and Energy, National Center for International Research on Photoelectric and Energy Materials, Yunnan University, Kunming650091, PR China
| |
Collapse
|
18
|
Wu J, Liu Y, Cao M, Zheng N, Ma H, Ye X, Yang N, Liu Z, Liao W, Sun L. Cancer-Responsive Multifunctional Nanoplatform Based on Peptide Self-Assembly for Highly Efficient Combined Cancer Therapy by Alleviating Hypoxia and Improving the Immunosuppressive Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:5667-5678. [PMID: 36651290 DOI: 10.1021/acsami.2c20388] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Hypoxia, as a main feature of the tumor microenvironment, has greatly limited the efficacy of photodynamic therapy (PDT), as well as its clinical application. Here, a multifunctional composite nanoplatform, the peptide/Ce6/MnO2 nanocomposite (RKCM), has been constructed to alleviate tumor hypoxia and increase the efficacy of PDT using rationally designed peptide fibrils to encapsulate chlorin e6 (Ce6) inside and to mineralize MnO2 nanoparticles on the surface. As a result, RKCM significantly improved the PDT efficacy by increasing reactive oxygen species (ROS) generation, decreasing tumor cell viability, and inhibiting tumor growth and metastasis. Besides, decreased HIF-1α expression and increased immune-activated cell infiltration were also observed in RKCM/laser treatment xenograft. Mechanically, (1) Ce6 can induce singlet oxygen (1O2) generation under laser irradiation to give photodynamic therapy (PDT); (2) MnO2 can react with H2O2 in situ to supply additional O2 to alleviate tumor hypoxia; and (3) the released Mn2+ ions can induce a Fenton-like reaction to generate •OH for chemical dynamic therapy (CDT). Moreover, RKCM/laser treatment also presented with an abscopal effect to block the occurrence of lung metastasis by remolding the pre-metastasis immune microenvironment. With these several aspects working together, the peptide/Ce6/MnO2 nanoplatform can achieve highly efficient tumor therapy. Such a strategy based on peptide self-assembly provides a promising way to rationally design a cancer-responsive multifunctional nanoplatform for highly efficient combined cancer therapy by alleviating hypoxia and improving the immune microenvironment.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Thoracic Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| | - Yang Liu
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Meiwen Cao
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Nannan Zheng
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Hongchao Ma
- State Key Laboratory of Heavy Oil Processing and Centre for Bioengineering and Biotechnology, College of Chemical Engineering, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Xiandong Ye
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Nanyan Yang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Zhihong Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Li Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Oncology, Air Force Medical Center of PLA, Air Force Medical University, Beijing 100089, China
| |
Collapse
|
19
|
Karballaei Mirzahosseini H, Sheikhi M, Najmeddin F, Shirangi M, Mojtahedzadeh M. 3D self-assembled nanocarriers for drug delivery. Drug Metab Rev 2023; 55:140-162. [PMID: 36772815 DOI: 10.1080/03602532.2023.2172182] [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: 07/14/2022] [Accepted: 01/19/2023] [Indexed: 02/12/2023]
Abstract
There are many benefits to drug delivery from drug-carrier nanostructure systems. It might be developed to carefully control drug release rates or to deliver a precise amount of a therapeutic substance to particular body areas. Self-assembling is the process by which molecules and nanoparticles spontaneously organize into organized clusters. For instance, proteins and peptides can interact with one another to create highly organized supramolecular structures with various properties, such as helical ribbons and fibrous scaffolds. Another advantage of self-assembly is that it may be effective with a variety of materials, including metals, oxides, inorganic salts, polymers, semiconductors, and even organic semiconductors. Fullerene, graphene, and carbon nanotubes (CNTs), three of the most fundamental classes of three-dimensionally self-assembling nanostructured carbon-based materials, are essential for the development of modern nanotechnologies. Self-assembled nanomaterials are used in a variety of fields, including nanotechnology, imaging, and biosensors. This review study begins with a summary of various major 3D nanomaterials, including graphene oxide, CNTs, and nanodiamond, as well as 3D self-assembled polyfunctionalized nanostructures and adaptable nanocarriers for drug delivery.
Collapse
Affiliation(s)
| | - Mojgan Sheikhi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Najmeddin
- Department of Clinical Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrnoosh Shirangi
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mojtaba Mojtahedzadeh
- Department of Clinical Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Pharmaceutical Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Anderson CF, Wang Q, Stern D, Leonard EK, Sun B, Fergie KJ, Choi CY, Spangler JB, Villano J, Pekosz A, Brayton CF, Jia H, Cui H. Supramolecular filaments for concurrent ACE2 docking and enzymatic activity silencing enable coronavirus capture and infection prevention. MATTER 2023; 6:583-604. [PMID: 36531610 PMCID: PMC9743467 DOI: 10.1016/j.matt.2022.11.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/11/2022] [Accepted: 11/16/2022] [Indexed: 06/01/2023]
Abstract
Coronaviruses have historically precipitated global pandemics of severe acute respiratory syndrome (SARS) into devastating public health crises. Despite the virus's rapid rate of mutation, all SARS coronavirus 2 (SARS-CoV-2) variants are known to gain entry into host cells primarily through complexation with angiotensin-converting enzyme 2 (ACE2). Although ACE2 has potential as a druggable decoy to block viral entry, its clinical use is complicated by its essential biological role as a carboxypeptidase and hindered by its structural and chemical instability. Here we designed supramolecular filaments, called fACE2, that can silence ACE2's enzymatic activity and immobilize ACE2 to their surface through enzyme-substrate complexation. This docking strategy enables ACE2 to be effectively delivered in inhalable aerosols and improves its structural stability and functional preservation. fACE2 exhibits enhanced and prolonged inhibition of viral entry compared with ACE2 alone while mitigating lung injury in vivo.
Collapse
Affiliation(s)
- Caleb F Anderson
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Qiong Wang
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David Stern
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Elissa K Leonard
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Boran Sun
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kyle J Fergie
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Chang-Yong Choi
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jamie B Spangler
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Jason Villano
- Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Cory F Brayton
- Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Hongpeng Jia
- Division of Pediatric Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
21
|
Wang Y, Su H, Wang Y, Cui H. Discovery of Y-Shaped Supramolecular Polymers in a Self-Assembling Peptide Amphiphile System. ACS Macro Lett 2022; 11:1355-1361. [PMID: 36413439 DOI: 10.1021/acsmacrolett.2c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Supramolecular polymers (SPs) formed by self-assembly of peptide-based molecular units assume a variety of interesting one-dimensional (1D) morphologies. While the morphological complexity and phase behavior of self-assembling peptide conjugates bear some resemblance to those of low-molecular-weight and macromolecular surfactants, Y-junctions, or three-way connected constructs, a topological defect observed in traditional surfactants has not been identified, likely due to the intolerance of defective packing by the strong, associative interactions afforded by the peptide segments. Here we report our discovery of branched SPs with Y-junctions and occasionally enlarged spherical end-caps formed by micellization of a ferrocene-based peptide amphiphile in water. Our results suggest that the incorporation of two ferrocenes into the amphiphile design is key to ensure the formation of branched SPs. We hypothesize that the complex interplay of internal interactions limits the effective propagation of hydrogen bonding within the assemblies and, consequently, creates fragmented β-sheets that are more tolerant for supramolecular branching. Given the redox sensitivity of the ferrocene units, sequential addition of reductants and oxidants to the solution led the assemblies to reversibly transform between branched SPs and spherical aggregates.
Collapse
Affiliation(s)
| | | | | | - Honggang Cui
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States.,Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, Maryland 21231, United States
| |
Collapse
|
22
|
Alnemeh-Al Ali H, Griveau A, Artzner F, Dupont A, Lautram N, Jourdain M, Eyer J. Investigation on the self-assembly of the NFL-TBS.40-63 peptide and its interaction with gold nanoparticles as a delivery agent for glioblastoma. Int J Pharm X 2022; 4:100128. [PMID: 36204592 PMCID: PMC9529584 DOI: 10.1016/j.ijpx.2022.100128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Affiliation(s)
| | - A. Griveau
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - F. Artzner
- Univ Rennes, CNRS, IPR (Institut de Physique de Rennes), UMR 6251, F-35000 Rennes, France
| | - A. Dupont
- Univ Rennes, CNRS, Inserm, BIOSIT-UMS 3480, US_S 018, Rennes, France
| | - N. Lautram
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - M.A. Jourdain
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
| | - J. Eyer
- Univ Angers, Inserm, CNRS, MINT, SFR ICAT, F-49000 Angers, France
- Corresponding author.
| |
Collapse
|
23
|
Wang L, Li C, Wang J, Yang G, Lv Y, Fu B, Jian L, Ma J, Yu J, Yang Z, Wu P, Li G, Liu X, Kang Z, Wang Z, Wang L, Wang H, Xu W. Transformable ECM Deprivation System Effectively Suppresses Renal Cell Carcinoma by Reversing Anoikis Resistance and Increasing Chemotherapy Sensitivity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2203518. [PMID: 36004775 DOI: 10.1002/adma.202203518] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Extracellular matrix (ECM) is crucial in various biological functions during tumor progression, including induction of anoikis resistance and cell adhesion-mediated drug resistance (CAM-DR). Fibronectin (FN) is a vital ECM component with direct regulatory effects on ECM-mediated anoikis resistance and CAM-DR, making it an attractive and innovative therapeutic target for depriving ECM in tumor tissue. Herein, an ECM deprivation system (EDS) is developed based on FN targeting self-assembly peptide for constructing nanofibers in the ECM of renal cell carcinoma (RCC), which contributes to: i) targeting and recognizing FN to form nanofibers for long-term retention in ECM, ii) reversing anoikis resistance via arresting the FN signaling pathway, and iii) serving as a drug-loading platform for sensitizing chemotherapy by ameliorating CAM-DR. The results reveal that EDS significantly reverses anoikis resistance of RCC cells by inhibiting the phosphorylation of FAK, a positive regulator of the FN signaling pathway. Meanwhile, EDS serves as a chemotherapy-sensitizer of cancer, exerting significant synergistic effects with doxorubicin (DOX). In vivo validation experiments show that EDS effectively suppresses metastasis and tumor growth with chemotherapy resistance. Collectively, the innovative EDS notably inhibits the tumor-promoting effect of ECM and may provide a novel approach for suppressing ECM and enhancing chemo-drug sensitivity.
Collapse
Affiliation(s)
- Lu Wang
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Cong Li
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Jiaqi Wang
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Guang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Yulin Lv
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Bo Fu
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Lingrui Jian
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Jinpeng Ma
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Jiaao Yu
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Zongzheng Yang
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Peng Wu
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Guangbin Li
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Xiao Liu
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Zhijian Kang
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Ziqi Wang
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| | - Lei Wang
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
- 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, 100190, 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, 100190, China
| | - Wanhai Xu
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
- Department of Urology, The Fourth Hospital of Harbin Medical University, Heilongjiang Key Laboratory of Scientific Research in Urology, No. 37 Yi-Yuan Street, Nangang District, Heilongjiang Province, Harbin, 150001, China
| |
Collapse
|
24
|
Mamidi N, García RG, Martínez JDH, Briones CM, Martínez Ramos AM, Tamez MFL, Del Valle BG, Segura FJM. Recent Advances in Designing Fibrous Biomaterials for the Domain of Biomedical, Clinical, and Environmental Applications. ACS Biomater Sci Eng 2022; 8:3690-3716. [PMID: 36037103 DOI: 10.1021/acsbiomaterials.2c00786] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Unique properties and potential applications of nanofibers have emerged as innovative approaches and opportunities in the biomedical, healthcare, environmental, and biosensor fields. Electrospinning and centrifugal spinning strategies have gained considerable attention among all kinds of strategies to produce nanofibers. These techniques produce nanofibers with high porosity and surface area, adequate pore architecture, and diverse chemical compositions. The extraordinary characteristics of nanofibers have unveiled new gates in nanomedicine to establish innovative fiber-based formulations for biomedical use, healthcare, and a wide range of other applications. The present review aims to provide a comprehensive overview of nanofibers and their broad range of applications, including drug delivery, biomedical scaffolds, tissue/bone-tissue engineering, dental applications, and environmental remediation in a single place. The review begins with a brief introduction followed by potential applications of nanofibers. Finally, the future perspectives and current challenges of nanofibers are demonstrated. This review will help researchers to engineer more efficient multifunctional nanofibers with improved characteristics for their effective use in broad areas. We strongly believe this review is a reader's delight and will help in dealing with the fundamental principles and applications of nanofiber-based scaffolds. This review will assist students and a broad range of scientific communities to understand the significance of nanofibers in several domains of nanotechnology, nanomedicine, biotechnology, and environmental remediation, which will set a benchmark for further research.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Rubén Gutiérrez García
- Department of Chemical Engineering, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64988, Mexico
| | - José Daniel Hernández Martínez
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Camila Martínez Briones
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Andrea Michelle Martínez Ramos
- Department of Biotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64988, Mexico
| | - María Fernanda Leal Tamez
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| | - Braulio González Del Valle
- Department of Chemical Engineering, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64988, Mexico
| | - Francisco Javier Macias Segura
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Monterrey, Nuevo Leon 64849, Mexico
| |
Collapse
|
25
|
Nanofiber Carriers of Therapeutic Load: Current Trends. Int J Mol Sci 2022; 23:ijms23158581. [PMID: 35955712 PMCID: PMC9368923 DOI: 10.3390/ijms23158581] [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: 06/30/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
The fast advancement in nanotechnology has prompted the improvement of numerous methods for the creation of various nanoscale composites of which nanofibers have gotten extensive consideration. Nanofibers are polymeric/composite fibers which have a nanoscale diameter. They vary in porous structure and have an extensive area. Material choice is of crucial importance for the assembly of nanofibers and their function as efficient drug and biomedicine carriers. A broad scope of active pharmaceutical ingredients can be incorporated within the nanofibers or bound to their surface. The ability to deliver small molecular drugs such as antibiotics or anticancer medications, proteins, peptides, cells, DNA and RNAs has led to the biomedical application in disease therapy and tissue engineering. Although nanofibers have shown incredible potential for drug and biomedicine applications, there are still difficulties which should be resolved before they can be utilized in clinical practice. This review intends to give an outline of the recent advances in nanofibers, contemplating the preparation methods, the therapeutic loading and release and the various therapeutic applications.
Collapse
|
26
|
Anderson CF, Chakroun RW, Grimmett ME, Domalewski CJ, Wang F, Cui H. Collagen-Binding Peptide-Enabled Supramolecular Hydrogel Design for Improved Organ Adhesion and Sprayable Therapeutic Delivery. NANO LETTERS 2022; 22:4182-4191. [PMID: 35522052 PMCID: PMC9844543 DOI: 10.1021/acs.nanolett.2c00967] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Spraying serves as an attractive, minimally invasive means of administering hydrogels for localized delivery, particularly due to high-throughput deposition of therapeutic depots over an entire target site of uneven surfaces. However, it remains a great challenge to design systems capable of rapid gelation after shear-thinning during spraying and adhering to coated tissues in wet, physiological environments. We report here on the use of a collagen-binding peptide to enable a supramolecular design of a biocompatible, bioadhesive, and sprayable hydrogel for sustained release of therapeutics. After spraying, the designed peptide amphiphile-based supramolecular filaments exhibit fast, physical cross-linking under physiological conditions. Our ex vivo studies suggest that the hydrogelator strongly adheres to the wet surfaces of multiple organs, and the extent of binding to collagen influences release kinetics from the gel. We envision that the sprayable organ-adhesive hydrogel can serve to enhance the efficacy of incorporated therapeutics for many biomedical applications.
Collapse
Affiliation(s)
- Caleb F Anderson
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Rami W Chakroun
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Maria E Grimmett
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Christopher J Domalewski
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Feihu Wang
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| |
Collapse
|
27
|
Panigrahi B, Singh RK, Suryakant U, Mishra S, Potnis AA, Jena AB, Kerry RG, Rajaram H, Ghosh SK, Mandal D. Cyclic peptides nanospheres: A '2-in-1' self-assembled delivery system for targeting nucleus and cytoplasm. Eur J Pharm Sci 2022; 171:106125. [PMID: 35033697 DOI: 10.1016/j.ejps.2022.106125] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 11/28/2022]
Abstract
Vascular endothelial growth factor (VEGF) is considered as one of the vital growth factors for angiogenesis, which is primarily responsible for the progress and maintenance of new vascular network in tumor. Numerous studies report that inhibition of VEGF-induced angiogenesis is a potent technique for cancer suppression. Recently, RNA interference, especially small interfering RNA (siRNA) signified a promising approach to suppress the gene expression. However, the clinical implementation of biological macromolecules such as siRNA is significantly limited because of stability and bioavailability issues. Herein, self-assembled peptide nanospheres have been generated from L,L-cyclic peptides using hydrophobic (Trp), positively charged (Arg) and cysteine (Cys) amino acid residues and demonstrated as vehicles for intracellular delivery of VEGF siRNA and VEGF antisense oligonucleotide. Formation of peptide nanostructures is confirmed by HR-TEM, AFM, SEM and DLS analysis. Possible mechanism of self-assembly of the cyclic peptides and their binding with macromolecules are demonstrated by in-silico analysis. Gel electrophoresis reveals that the newly generated peptide based organic materials exhibit strong binding affinity toward siRNAs / antisense oligonucleotides (ASOs) at optimum concentration. Flow cytometry and confocal microscopy results confirm the efficiency of the new biomaterials toward the intracellular delivery of fluorescent labeled siRNA / ASOs. Furthermore, VEGF expression evaluated by western blot and RT-PCR upon the delivery of functional VEGF siRNA/ASOs suggests that very low concentrations of VEGF siRNA/ASOs cause significant gene knockdown at protein and mRNA levels, respectively.
Collapse
Affiliation(s)
- Bijayananda Panigrahi
- School of Biotechnology, Kalinga Institute of Industrial Technology Deemed to be University, Campus 11, Patia, Bhubaneswar 751024, Odisha, India; Biopioneer Private Limited, Patia, Bhubaneswar, Odisha, India
| | - Rohit Kumar Singh
- School of Biotechnology, Kalinga Institute of Industrial Technology Deemed to be University, Campus 11, Patia, Bhubaneswar 751024, Odisha, India
| | - Uday Suryakant
- School of Biotechnology, Kalinga Institute of Industrial Technology Deemed to be University, Campus 11, Patia, Bhubaneswar 751024, Odisha, India
| | - Sourav Mishra
- School of Biotechnology, Kalinga Institute of Industrial Technology Deemed to be University, Campus 11, Patia, Bhubaneswar 751024, Odisha, India
| | - Akhilesh A Potnis
- Molecular Biology Division, Bhabha Atomic Research Centre (BARC), Trombay 400085, Mumbai, India
| | - Atala B Jena
- Centre Of Excellence In Integrated Omics and Computational Biology, Utkal University, Vani Vihar, Bhubaneswar, Odisha, 751004, India
| | - Rout George Kerry
- Department of Biotechnology, Utkal University, Vani Vihar, Bhubaneswar, Odisha 751004, India
| | - Hema Rajaram
- Molecular Biology Division, Bhabha Atomic Research Centre (BARC), Trombay 400085, Mumbai, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094 India
| | - Sunil K Ghosh
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094 India; Bio-Organic Division, Bhabha Atomic Research Centre (BARC), Trombay 400085, Mumbai India
| | - Dindyal Mandal
- School of Biotechnology, Kalinga Institute of Industrial Technology Deemed to be University, Campus 11, Patia, Bhubaneswar 751024, Odisha, India.
| |
Collapse
|
28
|
Paclitaxel Drug Delivery Systems: Focus on Nanocrystals' Surface Modifications. Polymers (Basel) 2022; 14:polym14040658. [PMID: 35215570 PMCID: PMC8875890 DOI: 10.3390/polym14040658] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 12/13/2022] Open
Abstract
Paclitaxel (PTX) is a chemotherapeutic agent that belongs to the taxane family and which was approved to treat various kinds of cancers including breast cancer, ovarian cancer, advanced non-small-cell lung cancer, and acquired immunodeficiency syndrome (AIDS)-related Kaposi’s sarcoma. Several delivery systems for PTX have been developed to enhance its solubility and pharmacological properties involving liposomes, nanoparticles, microparticles, micelles, cosolvent methods, and the complexation with cyclodextrins and other materials that are summarized in this article. Specifically, this review discusses deeply the developed paclitaxel nanocrystal formulations. As PTX is a hydrophobic drug with inferior water solubility properties, which are improved a lot by nanocrystal formulation. Based on that, many studies employed nano-crystallization techniques not only to improve the oral delivery of PTX, but IV, intraperitoneal (IP), and local and intertumoral delivery systems were also developed. Additionally, superior and interesting properties of PTX NCs were achieved by performing additional modifications to the NCs, such as stabilization with surfactants and coating with polymers. This review summarizes these delivery systems by shedding light on their route of administration, the methods used in the preparation and modifications, the in vitro or in vivo models used, and the advantages obtained based on the developed formulations.
Collapse
|
29
|
Xiang J, Liu X, Yuan G, Zhang R, Zhou Q, Xie T, Shen Y. Nanomedicine from amphiphilizedprodrugs: Concept and clinical translation. Adv Drug Deliv Rev 2021; 179:114027. [PMID: 34732344 DOI: 10.1016/j.addr.2021.114027] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/30/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
Nanomedicines generally consisting of carrier materials with small fractions of active pharmaceutical ingredients (API) have long been used to improve the pharmacokinetics and biodistributions, augment the therapeutic efficacies and mitigate the side effects. Amphiphilizing hydrophobic/hydrophilic drugs to prodrugs capable of self-assembly into well-defined nanostructures has emerged as a facile approach to fabricating nanomedicines because this amphiphilized prodrug (APD) strategy presents many advantages, including minimized use of inert carrier materials, well-characterized prodrug structures, fixed and high drug loading contents, 100% loading efficiency, and burst-free but controlled drug release. This review comprehensively summarizes recent advances in APDs and their nanomedicines, from the rationale and the stimuli-responsive linker chemistry for on-demand drug release to their progress to the clinics, clinical performance of APDs, as well as the challenges and perspective on future development.
Collapse
|
30
|
Pitz ME, Nukovic AM, Elpers MA, Alexander-Bryant AA. Factors Affecting Secondary and Supramolecular Structures of Self-Assembling Peptide Nanocarriers. Macromol Biosci 2021; 22:e2100347. [PMID: 34800001 DOI: 10.1002/mabi.202100347] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/19/2021] [Indexed: 01/12/2023]
Abstract
Self-assembling peptides are a popular vector for therapeutic cargo delivery due to their versatility, tunability, and biocompatibility. Accurately predicting secondary and supramolecular structures of self-assembling peptides is essential for de novo peptide design. However, computational modeling of such assemblies is not yet able to accurately predict structure formation for many peptide sequences. This review identifies patterns in literature between secondary and supramolecular structures, primary sequences, and applications to provide a guide for informed peptide design. An overview of peptide structures, their applications as nanocarriers, and analytical methods for characterizing secondary and supramolecular structure is examined. A top-down approach is then used to identify trends between peptide sequence and assembly structure from the current literature, including an analysis of the drivers at work, such as local and nonlocal sequence effects and solution conditions.
Collapse
Affiliation(s)
- Megan E Pitz
- Department of Bioengineering, 301 Rhodes Research Center, Clemson University, Clemson, SC, 29634-0905, USA
| | - Alexandra M Nukovic
- Department of Bioengineering, 301 Rhodes Research Center, Clemson University, Clemson, SC, 29634-0905, USA
| | - Margaret A Elpers
- Department of Bioengineering, 301 Rhodes Research Center, Clemson University, Clemson, SC, 29634-0905, USA
| | - Angela A Alexander-Bryant
- Department of Bioengineering, 301 Rhodes Research Center, Clemson University, Clemson, SC, 29634-0905, USA
| |
Collapse
|
31
|
Townsend PA, Kozhevnikova MV, Cexus ONF, Zamyatnin AA, Soond SM. BH3-mimetics: recent developments in cancer therapy. J Exp Clin Cancer Res 2021; 40:355. [PMID: 34753495 PMCID: PMC8576916 DOI: 10.1186/s13046-021-02157-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
The hopeful outcomes from 30 years of research in BH3-mimetics have indeed served a number of solid paradigms for targeting intermediates from the apoptosis pathway in a variety of diseased states. Not only have such rational approaches in drug design yielded several key therapeutics, such outputs have also offered insights into the integrated mechanistic aspects of basic and clinical research at the genetics level for the future. In no other area of medical research have the effects of such work been felt, than in cancer research, through targeting the BAX-Bcl-2 protein-protein interactions. With these promising outputs in mind, several mimetics, and their potential therapeutic applications, have also been developed for several other pathological conditions, such as cardiovascular disease and tissue fibrosis, thus highlighting the universal importance of the intrinsic arm of the apoptosis pathway and its input to general tissue homeostasis. Considering such recent developments, and in a field that has generated so much scientific interest, we take stock of how the broadening area of BH3-mimetics has developed and diversified, with a focus on their uses in single and combined cancer treatment regimens and recently explored therapeutic delivery methods that may aid the development of future therapeutics of this nature.
Collapse
Affiliation(s)
- Paul A Townsend
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
- University of Manchester, Manchester, UK.
| | - Maria V Kozhevnikova
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Andrey A Zamyatnin
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Lomonosov Moscow State University, Moscow, Russian Federation
- Sirius University of Science and Technology, Sochi, Russian Federation
| | - Surinder M Soond
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| |
Collapse
|
32
|
Shin MK, Mitrut R, Gu C, Kim LJ, Yeung BH, Lee R, Pham L, Tang WY, Sham JSK, Cui H, Polotsky VY. Pharmacological and Genetic Blockade of Trpm7 in the Carotid Body Treats Obesity-Induced Hypertension. Hypertension 2021; 78:104-114. [PMID: 33993722 PMCID: PMC8192446 DOI: 10.1161/hypertensionaha.120.16527] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Roxana Mitrut
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chenjuan Gu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bonnie H.Y. Yeung
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rachel Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luu Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wan-Yee Tang
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
| | - James S. K. Sham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Vsevolod Y. Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Mougin J, Bourgaux C, Couvreur P. Elongated self-assembled nanocarriers: From molecular organization to therapeutic applications. Adv Drug Deliv Rev 2021; 172:127-147. [PMID: 33705872 DOI: 10.1016/j.addr.2021.02.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/18/2020] [Accepted: 02/26/2021] [Indexed: 12/31/2022]
Abstract
Self-assembled cylindrical aggregates made of amphiphilic molecules emerged almost 40 years ago. Due to their length up to micrometers, those particles display original physico-chemical properties such as important flexibility and, for concentrated samples, a high viscoelasticity making them suitable for a wide range of industrial applications. However, a quarter of century was needed to successfully take advantage of those improvements towards therapeutic purposes. Since then, a wide diversity of biocompatible materials such as polymers, lipids or peptides, have been developed to design self-assembling elongated drug nanocarriers, suitable for therapeutic or diagnostic applications. More recently, the investigation of the main forces driving the unidirectional growth of these nanodevices allowed a translation toward the formation of pure nanodrugs to avoid the use of unnecessary side materials and the possible toxicity concerns associated.
Collapse
Affiliation(s)
- Julie Mougin
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry, France.
| | - Claudie Bourgaux
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry, France.
| | - Patrick Couvreur
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 92296 Châtenay-Malabry, France.
| |
Collapse
|
34
|
Synthesis and antibacterial study of cell-penetrating peptide conjugated trifluoroacetyl and thioacetyl lysine modified peptides. Eur J Med Chem 2021; 219:113447. [PMID: 33892275 DOI: 10.1016/j.ejmech.2021.113447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 11/23/2022]
Abstract
Substrate-based sirtuin inhibitors target bacterial genome and RNA and provide a promising approach to address bacterial resistance issues, if cellular internalisation can be achieved. We designed N-trifluoroacetyl lysine and N-thioacetyl lysine peptides (KP 13, KP 15 and KP 24) as inhibitors of bacterial sirtuins and their cell-penetrating peptide conjugates Tat KP 13, Tat KP 15 and Tat KP 24. The conjugated peptides were successfully internalised and showed signs of bacterial transcription inhibition resulting in enhanced antibacterial potency against model Gram negative and Gram positive pathogens. Synergistic activity in combination with streptomycin and polymyxin B has also been established. These peptides were effective in inhibiting biofilm formation and eradicating preformed biofilms. Morphological analysis using both SEM and TEM showed bacterial membrane disruption. Calcein dye leakage analysis established the selectivity of these peptides to bacterial membranes. This study documents the first report of the application of substrate-based sirtuin inhibitors as antimicrobial therapeutics.
Collapse
|
35
|
Kabir MT, Rahman MH, Akter R, Behl T, Kaushik D, Mittal V, Pandey P, Akhtar MF, Saleem A, Albadrani GM, Kamel M, Khalifa SA, El-Seedi HR, Abdel-Daim MM. Potential Role of Curcumin and Its Nanoformulations to Treat Various Types of Cancers. Biomolecules 2021; 11:392. [PMID: 33800000 PMCID: PMC8001478 DOI: 10.3390/biom11030392] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/27/2021] [Accepted: 03/03/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer is a major burden of disease globally. Each year, tens of millions of people are diagnosed with cancer worldwide, and more than half of the patients eventually die from it. Significant advances have been noticed in cancer treatment, but the mortality and incidence rates of cancers are still high. Thus, there is a growing research interest in developing more effective and less toxic cancer treatment approaches. Curcumin (CUR), the major active component of turmeric (Curcuma longa L.), has gained great research interest as an antioxidant, anticancer, and anti-inflammatory agent. This natural compound shows its anticancer effect through several pathways including interfering with multiple cellular mechanisms and inhibiting/inducing the generation of multiple cytokines, enzymes, or growth factors including IκB kinase β (IκKβ), tumor necrosis factor-alpha (TNF-α), signal transducer, and activator of transcription 3 (STAT3), cyclooxygenase II (COX-2), protein kinase D1 (PKD1), nuclear factor-kappa B (NF-κB), epidermal growth factor, and mitogen-activated protein kinase (MAPK). Interestingly, the anticancer activity of CUR has been limited primarily due to its poor water solubility, which can lead to low chemical stability, low oral bioavailability, and low cellular uptake. Delivering drugs at a controlled rate, slow delivery, and targeted delivery are other very attractive methods and have been pursued vigorously. Multiple CUR nanoformulations have also been developed so far to ameliorate solubility and bioavailability of CUR and to provide protection to CUR against hydrolysis inactivation. In this review, we have summarized the anticancer activity of CUR against several cancers, for example, gastrointestinal, head and neck, brain, pancreatic, colorectal, breast, and prostate cancers. In addition, we have also focused on the findings obtained from multiple experimental and clinical studies regarding the anticancer effect of CUR in animal models, human subjects, and cancer cell lines.
Collapse
Affiliation(s)
- Md. Tanvir Kabir
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka 1212, Bangladesh;
| | - Md. Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka 1100, Bangladesh;
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India; (D.K.); (V.M.)
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak 124001, India; (D.K.); (V.M.)
| | - Parijat Pandey
- Shri Baba Mastnath Institute of Pharmaceutical Sciences and Research, Baba Mastnath University, Rohtak 124001, India;
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Lahore Campus, Riphah International University, Lahore 54000, Pakistan;
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Shaden A.M. Khalifa
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, S-10691 Stockholm, Sweden
| | - Hesham R. El-Seedi
- Pharmacognosy Group, Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Box 574, 751 23 Uppsala, Sweden;
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Kom 32512, Egypt
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
36
|
Moncho-Jordá A, Jódar-Reyes AB, Kanduč M, Germán-Bellod A, López-Romero JM, Contreras-Cáceres R, Sarabia F, García-Castro M, Pérez-Ramírez HA, Odriozola G. Scaling Laws in the Diffusive Release of Neutral Cargo from Hollow Hydrogel Nanoparticles: Paclitaxel-Loaded Poly(4-vinylpyridine). ACS NANO 2020; 14:15227-15240. [PMID: 33174725 DOI: 10.1021/acsnano.0c05480] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We study the nonequilibrium diffusive release of electroneutral molecular cargo encapsulated inside hollow hydrogel nanoparticles. We propose a theoretical model that includes osmotic, steric, and short-range polymer-cargo attractions to determine the effective cargo-hydrogel interaction, ueff*, and the effective diffusion coefficient of the cargo inside the polymer network, Deff*. Using dynamical density functional theory (DDFT), we investigate the scaling of the characteristic release time, τ1/2, with the key parameters involved in the process, namely, ueff*, Deff*, and the swelling ratio. This effort represents a full study of the problem, covering a broad range of cargo sizes and providing predictions for repulsive and attractive polymer shells. Our calculations show that the release time through repulsive polymer networks scales with q2eβueff*/Deff* for βueff* ≫ 1. In this case, the cargo molecules are excluded from the shell of the hydrogel. For attractive shells, the polymer retains the cargo molecules on its internal surface and its interior, and the release time grows exponentially with the attraction strength. The DDFT calculations are compared to an analytical model for the mean first passage time, which provides an excellent quantitative description of the kinetics for both repulsive and attractive shells without fitting parameters. Finally, we apply the method to reproduce experimental results on the release of paclitaxel from hollow poly(4-vinylpyridine) nanoparticles and find that the slow release of the drug can be explained in terms of the strong binding attraction between the drug and the polymer.
Collapse
Affiliation(s)
- Arturo Moncho-Jordá
- Departamento de Física Aplicada, Universidad de Granada, Campus Fuentenueva S/N, 18071 Granada, Spain
- Instituto Carlos I de Física Teórica y Computacional, Facultad de Ciencias, Universidad de Granada, Campus Fuentenueva S/N, 18071 Granada, Spain
| | - Ana B Jódar-Reyes
- Departamento de Física Aplicada, Universidad de Granada, Campus Fuentenueva S/N, 18071 Granada, Spain
- Excellence Research Unit "Modeling Nature" (MNat), Universidad de Granada, Campus Fuentenueva S/N, 18071 Granada, Spain
| | - Matej Kanduč
- Jožef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Alicia Germán-Bellod
- Departamento de Física Aplicada, Universidad de Granada, Campus Fuentenueva S/N, 18071 Granada, Spain
| | - Juan M López-Romero
- Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain
| | - Rafael Contreras-Cáceres
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, 28040 Madrid, Spain
| | - Francisco Sarabia
- Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain
| | - Miguel García-Castro
- Departamento de Química Orgánica, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga, Spain
| | - Héctor A Pérez-Ramírez
- Física de Procesos Irreversibles, Ciencias Básicas e Ingeniería, Universidad Autónoma Metropolitana-Azcapotzalco, Avenida San Pablo 180, 02200 Ciudad de México, Mexico
| | - Gerardo Odriozola
- Física de Procesos Irreversibles, Ciencias Básicas e Ingeniería, Universidad Autónoma Metropolitana-Azcapotzalco, Avenida San Pablo 180, 02200 Ciudad de México, Mexico
| |
Collapse
|
37
|
Yin X, Chen Z, Chen Y, Xie Y, Xiong B, Jiang H, Zhu J. Lipidated gemini peptide amphiphiles with enhanced loading capacity and cell membrane affinity for drug delivery. Colloids Surf B Biointerfaces 2020; 195:111271. [DOI: 10.1016/j.colsurfb.2020.111271] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/17/2022]
|
38
|
Peng F, Zhang W, Qiu F. Self-assembling Peptides in Current Nanomedicine: Versatile Nanomaterials for Drug Delivery. Curr Med Chem 2020; 27:4855-4881. [PMID: 31309877 DOI: 10.2174/0929867326666190712154021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/27/2019] [Accepted: 06/11/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The development of modern nanomedicine greatly depends on the involvement of novel materials as drug delivery system. In order to maximize the therapeutic effects of drugs and minimize their side effects, a number of natural or synthetic materials have been widely investigated for drug delivery. Among these materials, biomimetic self-assembling peptides (SAPs) have received more attention in recent years. Considering the rapidly growing number of SAPs designed for drug delivery, a summary of how SAPs-based drug delivery systems were designed, would be beneficial. METHOD We outlined research works on different SAPs that have been investigated as carriers for different drugs, focusing on the design of SAPs nanomaterials and how they were used for drug delivery in different strategies. RESULTS Based on the principle rules of chemical complementarity and structural compatibility, SAPs such as ionic self-complementary peptide, peptide amphiphile and surfactant-like peptide could be designed. Determined by the features of peptide materials and the drugs to be delivered, different strategies such as hydrogel embedding, hydrophobic interaction, electrostatic interaction, covalent conjugation or the combination of them could be employed to fabricate SAPs-drug complex, which could achieve slow release, targeted or environment-responsive delivery of drugs. Furthermore, some SAPs could also be combined with other types of materials for drug delivery, or even act as drug by themselves. CONCLUSION Various types of SAPs have been designed and used for drug delivery following various strategies, suggesting that SAPs as a category of versatile nanomaterials have promising potential in the field of nanomedicine.
Collapse
Affiliation(s)
- Fei Peng
- Laboratory of Anaesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wensheng Zhang
- Laboratory of Anaesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Feng Qiu
- Laboratory of Anaesthesia and Critical Care Medicine, Translational Neuroscience Centre, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
39
|
Madhanagopal B, More SH, Bansode ND, Ganesh KN. Conformation and Morphology of 4-(NH 2/OH)-Substituted l/d-Prolyl Polypeptides: Effect of Homo- and Heterochiral Backbones on Formation of β-Structures and Nanofibers. ACS OMEGA 2020; 5:21781-21795. [PMID: 32905392 PMCID: PMC7469381 DOI: 10.1021/acsomega.0c02826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/04/2020] [Indexed: 06/11/2023]
Abstract
The relative stereochemistry of C2 and C4 in 4-substituted prolyl polypeptides plays an important role in defining the derived conformation in solution. cis-(2S,4S)-Amino/hydroxy-l-prolyl polypeptide (lC-Amp 9/lC-Hyp 9) shows a PPII conformation in phosphate buffer and a β-structure in a relatively hydrophobic solvent, trifluoroethanol (TFE). It is now demonstrated that the homochiral enantiomeric cis-substituted d-prolyl polypeptide (dC-Amp 9/dC-Hyp 9) exhibits mirror image β-structures in TFE. In the case of alternating heterochiral prolyl peptides, it is the trans-substituted [lT(2S,4R)-dT(2R,4S)] n prolyl polypeptide that shows β-structures in TFE, while the cis-substituted [lC(2S,4S)-dC(2R,4R)] n prolyl polypeptide is disordered in both phosphate buffer and TFE. The results highlight the important chirality-specific structural requirements for β-structure formation. The observed conformation in solution (circular dichroism (CD)) is also correlated with the morphology of the self-assemblies (field emission scanning electron microscopy (FESEM)), with the PPII form leading to spherical nanoparticles and β-structures leading to nanofiber formation. The results shed light on the role of relative stereochemistry at C2 and C4 in defining the polyproline peptide conformation in solution and how different conformations drive self-assemblies of peptides toward specific nanostructures.
Collapse
Affiliation(s)
- Bharath
Raj Madhanagopal
- Indian
Institute of Science Education and Research (IISER), Tirupati, Karkambadi Road, Tirupati 517507, Andhra Pradesh, India
| | - Shahaji H. More
- Indian
Institute of Science Education and Research (IISER), Tirupati, Karkambadi Road, Tirupati 517507, Andhra Pradesh, India
| | - Nitin D. Bansode
- LCPO,
ENSCBP, UMR 5629, University of Bordeaux, Pessac 33600, France
| | - Krishna N. Ganesh
- Indian
Institute of Science Education and Research (IISER), Tirupati, Karkambadi Road, Tirupati 517507, Andhra Pradesh, India
- Indian
Institute of Science Education and Research (IISER), Pune, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
| |
Collapse
|
40
|
Wang F, Su H, Lin R, Chakroun RW, Monroe MK, Wang Z, Porter M, Cui H. Supramolecular Tubustecan Hydrogel as Chemotherapeutic Carrier to Improve Tumor Penetration and Local Treatment Efficacy. ACS NANO 2020; 14:10083-10094. [PMID: 32806082 DOI: 10.1021/acsnano.0c03286] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Local chemotherapy is a clinically proven strategy in treating malignant brain tumors. Its benefits, however, are largely limited by the rapid release and clearance of therapeutic agents and the inability to penetrate through tumor tissues. We report here on a supramolecular tubustecan (TT) hydrogel as both a therapeutic and drug carrier that enables long-term, sustained drug release and improved tumor tissue penetration. Covalent linkage of a tissue penetrating cyclic peptide to two camptothecin drug units creates a TT prodrug amphiphile that can associate into tubular supramolecular polymers and subsequently form a well-defined sphere-shaped hydrogel after injection into tumor tissues. The hollow nature of the resultant tubular assemblies allows for encapsulation of doxorubicin or curcumin for combination therapy. Our in vitro and in vivo studies reveal that these dual drug-bearing supramolecular hydrogels enhance tumor retention and penetration, serving as a local therapeutic depot for potent tumor regression, inhibition of tumor metastasis and recurrence, and mitigation of the off-target side effects.
Collapse
Affiliation(s)
- Feihu Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Hao Su
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Ran Lin
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Rami W Chakroun
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Maya K Monroe
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Zongyuan Wang
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Michael Porter
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Institute for NanoBiotechnology (INBT), Johns Hopkins University, Baltimore, Maryland 21218, United States
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| |
Collapse
|
41
|
Abstract
Cell-penetrating peptides present huge biomedical applications in a variety of pathologies, thanks to their ability to penetrate membranes and carry a variety of cargoes inside cells. Progress in peptide synthesis has produced a greater availability of virtually any synthetic peptide, increasing their attractiveness. Most molecules when associated to a cell-penetrating peptides can be delivered into a cell, however, understanding of the critical factors influencing the uptake mechanism is of paramount importance to construct nanoplatforms for effective delivery in vitro and in vivo in medical applications. Focus is now on the state-of-art of the mechanisms enabling therapeutics/diagnostics to reach the site target of their activities, and in support of scientists developing platforms for drug delivery and personalized therapies.
Collapse
|
42
|
Wang Z, Chen J, Little N, Lu J. Self-assembling prodrug nanotherapeutics for synergistic tumor targeted drug delivery. Acta Biomater 2020; 111:20-28. [PMID: 32454086 PMCID: PMC7245299 DOI: 10.1016/j.actbio.2020.05.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/27/2020] [Accepted: 05/18/2020] [Indexed: 01/08/2023]
Abstract
Self-assembling prodrugs represents a robust and effective nanotherapeutic approach for delivering poorly soluble anticancer drugs. With numerous intrinsic advantages, self-assembling prodrugs possess the maximum drug loading capacity, controlled drug release kinetics, prolonged blood circulation, and preferential tumor accumulation based on the enhanced permeability and retention (EPR) effect. These prodrug conjugates allow for efficient self-assembly into nanodrugs with the potential of encapsulating other therapeutic agents that have different molecular targets, enabling simultaneous temporal-spatial release of drugs for synergistic antitumor efficacy with reduced systemic side effects. The aim of this review is to summarize the recent progress of self-assembling prodrug cancer nanotherapeutics that are made through conjugating therapeutically active agents to Polyethylene glycol, Vitamin E, or drugs with different physicochemical properties via rational design, for synergistic tumor targeted drug delivery. Statement of Significance All current FDA-approved nanomedicines use inert biomaterials as drug delivery carriers. These biomaterials lack any therapeutic potential, contributing not only to the cost, but may also elicit severe unfavorable adverse effects. Despite the reduction in toxicity associated with the payload, these nanotherapeutics have been met with limited clinical success, likely due to the monotherapy regimen. The self-assembling prodrug (SAP) has been emerging as a powerful platform for enhancing efficacy through co-delivering other therapeutic modalities with distinct molecular targets. Herein, we opportunely present a comprehensive review article summarizing three unique approaches of making SAP for synergistic drug delivery: pegylation, vitamin E-derivatization, and drug-drug conjugation. These SAPs may inevitably pave the way for developing more efficacious, clinically translatable, combination cancer nanotherapies.
Collapse
|
43
|
Adibnia V, Mirbagheri M, Salimi S, De Crescenzo G, Banquy X. Nonspecific interactions in biomedical applications. Curr Opin Colloid Interface Sci 2020. [DOI: 10.1016/j.cocis.2019.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Liu Y, Wang Q, Lu Y, Deng H, Zhou X. Synergistic enhancement of cytotoxicity against cancer cells by incorporation of rectorite into the paclitaxel immobilized cellulose acetate nanofibers. Int J Biol Macromol 2020; 152:672-680. [DOI: 10.1016/j.ijbiomac.2020.02.184] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/15/2020] [Accepted: 02/16/2020] [Indexed: 12/18/2022]
|
45
|
Lee S, Pham TC, Bae C, Choi Y, Kim YK, Yoon J. Nano theranostics platforms that utilize proteins. Coord Chem Rev 2020. [DOI: 10.1016/j.ccr.2020.213258] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
46
|
Su H, Wang F, Ran W, Zhang W, Dai W, Wang H, Anderson CF, Wang Z, Zheng C, Zhang P, Li Y, Cui H. The role of critical micellization concentration in efficacy and toxicity of supramolecular polymers. Proc Natl Acad Sci U S A 2020; 117:4518-4526. [PMID: 32071209 PMCID: PMC7060728 DOI: 10.1073/pnas.1913655117] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The inception and development of supramolecular chemistry have provided a vast library of supramolecular structures and materials for improved practice of medicine. In the context of therapeutic delivery, while supramolecular nanostructures offer a wide variety of morphologies as drug carriers for optimized targeting and controlled release, concerns are often raised as to how their morphological stability and structural integrity impact their in vivo performance. After intravenous (i.v.) administration, the intrinsic reversible and dynamic feature of supramolecular assemblies may lead them to dissociate upon plasma dilution to a concentration below their critical micellization concentration (CMC). As such, CMC represents an important characteristic for supramolecular biomaterials design, but its pharmaceutical role remains elusive. Here, we report the design of a series of self-assembling prodrugs (SAPDs) that spontaneously associate in aqueous solution into supramolecular polymers (SPs) with varying CMCs. Two hydrophobic camptothecin (CPT) molecules were conjugated onto oligoethylene-glycol (OEG)-decorated segments with various OEG repeat numbers (2, 4, 6, 8). Our studies show that the lower the CMC, the lower the maximum tolerated dose (MTD) in rodents. When administrated at the same dosage of 10 mg/kg (CPT equivalent), SAPD 1, the one with the lowest CMC, shows the best efficacy in tumor suppression. These observations can be explained by the circulation and dissociation of SAPD SPs and the difference in molecular and supramolecular distribution between excretion and organ uptake. We believe these findings offer important insight into the role of supramolecular stability in determining their therapeutic index and in vivo efficacy.
Collapse
Affiliation(s)
- Hao Su
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
| | - Feihu Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
| | - Wei Ran
- State Key Laboratory of Drug Research and Center for Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Weijie Zhang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wenbing Dai
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Han Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
| | - Caleb F Anderson
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
| | - Zongyuan Wang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
- Collaborative Innovation Center of Chemical Science and Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Chao Zheng
- State Key Laboratory of Drug Research and Center for Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research and Center for Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center for Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218;
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218
- Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
47
|
Taylor RE, Zahid M. Cell Penetrating Peptides, Novel Vectors for Gene Therapy. Pharmaceutics 2020; 12:E225. [PMID: 32138146 PMCID: PMC7150854 DOI: 10.3390/pharmaceutics12030225] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/27/2020] [Accepted: 03/01/2020] [Indexed: 12/31/2022] Open
Abstract
Cell penetrating peptides (CPPs), also known as protein transduction domains (PTDs), first identified ~25 years ago, are small, 6-30 amino acid long, synthetic, or naturally occurring peptides, able to carry variety of cargoes across the cellular membranes in an intact, functional form. Since their initial description and characterization, the field of cell penetrating peptides as vectors has exploded. The cargoes they can deliver range from other small peptides, full-length proteins, nucleic acids including RNA and DNA, liposomes, nanoparticles, and viral particles as well as radioisotopes and other fluorescent probes for imaging purposes. In this review, we will focus briefly on their history, classification system, and mechanism of transduction followed by a summary of the existing literature on use of CPPs as gene delivery vectors either in the form of modified viruses, plasmid DNA, small interfering RNA, oligonucleotides, full-length genes, DNA origami or peptide nucleic acids.
Collapse
Affiliation(s)
- Rebecca E. Taylor
- Mechanical Engineering, Biomedical Engineering and Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA;
| | - Maliha Zahid
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, USA
| |
Collapse
|
48
|
Self-assembling mertansine prodrug improves tolerability and efficacy of chemotherapy against metastatic triple-negative breast cancer. J Control Release 2020; 318:234-245. [DOI: 10.1016/j.jconrel.2019.12.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 12/04/2019] [Accepted: 12/15/2019] [Indexed: 12/11/2022]
|
49
|
Cai Y, Ran W, Zhai Y, Wang J, Zheng C, Li Y, Zhang P. Recent progress in supramolecular peptide assemblies as virus mimics for cancer immunotherapy. Biomater Sci 2020; 8:1045-1057. [DOI: 10.1039/c9bm01380f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Supramolecular peptide assemblies can mimic natural viruses and serve as well-defined, dynamic and multifunctional nanoplatforms for cancer immunotherapy, where the peptide segments act as antigens, adjuvants and carriers.
Collapse
Affiliation(s)
- Ying Cai
- State Key Laboratory of Drug Research & Center of Pharmaceutics
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Wei Ran
- State Key Laboratory of Drug Research & Center of Pharmaceutics
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Yihui Zhai
- State Key Laboratory of Drug Research & Center of Pharmaceutics
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Junyang Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Chao Zheng
- State Key Laboratory of Drug Research & Center of Pharmaceutics
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research & Center of Pharmaceutics
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| |
Collapse
|
50
|
Kiran S, Dwivedi P, Khatik R, Hameed S, Dwivedi M, Huang F, Xu RX. Synthesis of a functionalized dipeptide for targeted delivery and pH-sensitive release of chemotherapeutics. Chem Commun (Camb) 2020; 56:285-288. [DOI: 10.1039/c9cc09131a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Design of pH-sensitive folic acid conjugated diphenyl peptide nanoparticles for targeted folate receptors mediated endocytosis.
Collapse
Affiliation(s)
- Sonia Kiran
- Hefei National Laboratory of Physical Sciences at Microscale
- Department of Chemistry
- University of Science and Technology of China
- Hefei
- China
| | - Pankaj Dwivedi
- School of Engineering Science
- Department of Precision Machinery and Precision Instrumentation
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Renuka Khatik
- Hefei National Laboratory of Physical Sciences at Microscale
- Department of Chemistry
- University of Science and Technology of China
- Hefei
- China
| | - Sadaf Hameed
- Department of Biomedical Engineering
- College of Engineering
- Peking University
- Beijing 100871
- China
| | - Monika Dwivedi
- School of Engineering Science
- Department of Precision Machinery and Precision Instrumentation
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Fangsheng Huang
- School of Engineering Science
- Department of Precision Machinery and Precision Instrumentation
- University of Science and Technology of China
- Hefei
- P. R. China
| | - Ronald X. Xu
- School of Engineering Science
- Department of Precision Machinery and Precision Instrumentation
- University of Science and Technology of China
- Hefei
- P. R. China
| |
Collapse
|