1
|
Khanolkar B, Shende P. BSA nanoclusters-based sensor for detection of dopamine in schizophrenia from biofluids. Drug Dev Ind Pharm 2024; 50:341-353. [PMID: 38470160 DOI: 10.1080/03639045.2024.2328722] [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: 09/19/2023] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
OBJECTIVE To develop nontoxic and stable fluorescent emission B-Cu nanoclusters (NCs) for the specific detection of dopamine at low concentrations in cerebrospinal fluid (CSF). SIGNIFICANCE Fluorescent gold and copper NCs conjugated with proteins, such as bovine serum albumin (BSA), offer photostability and healthcare potential. This study focused on fabricating B-Cu NCs that exhibited superior characteristics for sensitive dopamine detection. METHODS The study employed various instrumental techniques including attenuated total reflectance Fourier-transform infrared spectroscopy (ATR-FTIR), scanning electron microscopy (SEM), spectrofluorometry, and transmission electron microscopy (TEM) to characterize the formulated B-Cu NCs. The NCs were synthesized, resulting in particle size ∼300 nm. The highest observed fluorescence was recorded at 24542.81 relative fluorescence units (RFU). RESULTS The introduction of dopamine at concentrations of 0.1, 0.2, 0.3, and 0.4 ng/mL led to decreased fluorescence in both B-Au and B-Cu NCs due to an electron transport system. This reduction in fluorescence allowed dopamine concentration analysis in phosphate buffer and biological fluids such as blood plasma and CSF. B-Cu NCs showed potential as a biosensing system for point-of-care (POC) applications, specifically for diagnosing schizophrenia. CONCLUSION The study successfully synthesized stable and nontoxic B-Cu NCs with enhanced fluorescent emission properties. These NCs exhibited the capacity to detect dopamine at low concentrations in CSF. The study's findings hold promise for future applications, particularly in the development of a B-Cu NCs-based biosensing system for convenient POC detection of schizophrenia by both patients and clinicians. The potential impact of this technology on healthcare and biomedical fields is substantial.
Collapse
Affiliation(s)
- Bhakti Khanolkar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, India
| | - Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Mumbai, India
| |
Collapse
|
2
|
Yang N, Sun M, Wang H, Hu D, Zhang A, Khan S, Chen Z, Chen D, Xie S. Progress of stimulus responsive nanosystems for targeting treatment of bacterial infectious diseases. Adv Colloid Interface Sci 2024; 324:103078. [PMID: 38215562 DOI: 10.1016/j.cis.2024.103078] [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: 08/17/2023] [Revised: 12/27/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
In recent decades, due to insufficient concentration at the lesion site, low bioavailability and increasingly serious resistance, antibiotics have become less and less dominant in the treatment of bacterial infectious diseases. It promotes the development of efficient drug delivery systems, and is expected to achieve high absorption, targeted drug release and satisfactory therapy effects. A variety of endogenous stimulation-responsive nanosystems have been constructed by using special infection microenvironments (pH, enzymes, temperature, etc.). In this review, we firstly provide an extensive review of the current research progress in antibiotic treatment dilemmas and drug delivery systems. Then, the mechanism of microenvironment characteristics of bacterial infected lesions was elucidated to provide a strong theoretical basis for bacteria-targeting nanosystems design. In particular, the discussion focuses on the design principles of single-stimulus and dual-stimulus responsive nanosystems, as well as the use of endogenous stimulus-responsive nanosystems to deliver antimicrobial agents to target locations for combating bacterial infectious diseases. Finally, the challenges and prospects of endogenous stimulus-responsive nanosystems were summarized.
Collapse
Affiliation(s)
- Niuniu Yang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Mengyuan Sun
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Huixin Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Danlei Hu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Aoxue Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Suliman Khan
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Zhen Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China
| | - Dongmei Chen
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Shuyu Xie
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei 430070, China; Shenzhen Institute of Nutrition and Health,Huazhong Agricultural University, Shenzhen, China; Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| |
Collapse
|
3
|
Mahajan K, Bhattacharya S. The Advancement and Obstacles in Improving the Stability of Nanocarriers for Precision Drug Delivery in the Field of Nanomedicine. Curr Top Med Chem 2024; 24:686-721. [PMID: 38409730 DOI: 10.2174/0115680266287101240214071718] [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: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
Nanocarriers have emerged as a promising class of nanoscale materials in the fields of drug delivery and biomedical applications. Their unique properties, such as high surface area- tovolume ratios and enhanced permeability and retention effects, enable targeted delivery of therapeutic agents to specific tissues or cells. However, the inherent instability of nanocarriers poses significant challenges to their successful application. This review highlights the importance of nanocarrier stability in biomedical applications and its impact on biocompatibility, targeted drug delivery, long shelf life, drug delivery performance, therapeutic efficacy, reduced side effects, prolonged circulation time, and targeted delivery. Enhancing nanocarrier stability requires careful design, engineering, and optimization of physical and chemical parameters. Various strategies and cutting-edge techniques employed to improve nanocarrier stability are explored, with a focus on their applications in drug delivery. By understanding the advances and challenges in nanocarrier stability, this review aims to contribute to the development and implementation of nanocarrier- based therapies in clinical settings, advancing the field of nanomedicine.
Collapse
Affiliation(s)
- Kalpesh Mahajan
- Department of Quality Assurence, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKMS NMIMS Maharashtra, Shirpur, 425405, India
| |
Collapse
|
4
|
Yao L, Liu Q, Lei Z, Sun T. Development and challenges of antimicrobial peptide delivery strategies in bacterial therapy: A review. Int J Biol Macromol 2023; 253:126819. [PMID: 37709236 DOI: 10.1016/j.ijbiomac.2023.126819] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023]
Abstract
The escalating global prevalence of antimicrobial resistance poses a critical threat, prompting concerns about its impact on public health. This predicament is exacerbated by the acute shortage of novel antimicrobial agents, a scarcity attributed to the rapid surge in bacterial resistance. This review delves into the realm of antimicrobial peptides, a diverse class of compounds ubiquitously present in plants and animals across various natural organisms. Renowned for their intrinsic antibacterial activity, these peptides provide a promising avenue to tackle the intricate challenge of bacterial resistance. However, the clinical utility of peptide-based drugs is hindered by limited bioavailability and susceptibility to rapid degradation, constraining efforts to enhance the efficacy of bacterial infection treatments. The emergence of nanocarriers marks a transformative approach poised to revolutionize peptide delivery strategies. This review elucidates a promising framework involving nanocarriers within the realm of antimicrobial peptides. This paradigm enables meticulous and controlled peptide release at infection sites by detecting dynamic shifts in microenvironmental factors, including pH, ROS, GSH, and reactive enzymes. Furthermore, a glimpse into the future reveals the potential of targeted delivery mechanisms, harnessing inflammatory responses and intricate signaling pathways, including adenosine triphosphate, macrophage receptors, and pathogenic nucleic acid entities. This approach holds promise in fortifying immunity, thereby amplifying the potency of peptide-based treatments. In summary, this review spotlights peptide nanosystems as prospective solutions for combating bacterial infections. By bridging antimicrobial peptides with advanced nanomedicine, a new therapeutic era emerges, poised to confront the formidable challenge of antimicrobial resistance head-on.
Collapse
Affiliation(s)
- Longfukang Yao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China
| | - Qianying Liu
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China; Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, Wuhan University of Technology, 122 Luoshi Road, Wuhan 430070, China.
| |
Collapse
|
5
|
Biomaterial therapeutic strategies for treatment of bacterial lung infections. Biofilm 2023; 5:100111. [PMID: 36909663 PMCID: PMC9999167 DOI: 10.1016/j.bioflm.2023.100111] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Bacterial infections of the lung frequently occur as a secondary infection to many respiratory viral infections and conditions, including influenza, COVID-19, chronic obstructive pulmonary disease (COPD), and cystic fibrosis (CF). Currently, clinical standard treats bacterial infections of the lung with antibiotic drugs. However, the use of broad-spectrum antibiotics can disrupt host microbiomes, lead to patient discomfort, and current clinical settings face the constantly increasing threat of drug-resistant bacteria. Biofilms further obstruct effective treatment due to their protective matrix layer, which shields bacteria from both the host immune system and antimicrobial drugs and subsequently promotes drug resistance. Alternative antimicrobial agents, including bacteriophages and antimicrobial peptides, have been utilized to treat drug-resistant bacteria. However, these antimicrobial agents have significant limitations pertaining to their ability to arrive at infection sites without compromised function and ability to persist over an extended period to fully treat infections. Enhanced delivery strategies present great promise in addressing these issues by using micro/nanoparticle carriers that shield antimicrobial agents in transit and result in sustained release, enhancing subsequent therapeutic effect and can even be modulated to be multi-functional to further improve recovery following bacterial infection.
Collapse
|
6
|
Colilla M, Vallet-Regí M. Organically Modified Mesoporous Silica Nanoparticles against Bacterial Resistance. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2023; 35:8788-8805. [PMID: 38027542 PMCID: PMC10653088 DOI: 10.1021/acs.chemmater.3c02192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/22/2023] [Indexed: 12/01/2023]
Abstract
Bacterial antimicrobial resistance is posed to become a major hazard to global health in the 21st century. An aggravating issue is the stalled antibiotic research pipeline, which requires the development of new therapeutic strategies to combat antibiotic-resistant infections. Nanotechnology has entered into this scenario bringing up the opportunity to use nanocarriers capable of transporting and delivering antimicrobials to the target site, overcoming bacterial resistant barriers. Among them, mesoporous silica nanoparticles (MSNs) are receiving growing attention due to their unique features, including large drug loading capacity, biocompatibility, tunable pore sizes and volumes, and functionalizable silanol-rich surface. This perspective article outlines the recent research advances in the design and development of organically modified MSNs to fight bacterial infections. First, a brief introduction to the different mechanisms of bacterial resistance is presented. Then, we review the recent scientific approaches to engineer multifunctional MSNs conceived as an assembly of inorganic and organic building blocks, against bacterial resistance. These elements include specific ligands to target planktonic bacteria, intracellular bacteria, or bacterial biofilm; stimuli-responsive entities to prevent antimicrobial cargo release before arriving at the target; imaging agents for diagnosis; additional constituents for synergistic combination antimicrobial therapies; and aims to improve the therapeutic outcomes. Finally, this manuscript addresses the current challenges and future perspectives on this hot research area.
Collapse
Affiliation(s)
- Montserrat Colilla
- Departamento
de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación
Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| | - María Vallet-Regí
- Departamento
de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación
Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
7
|
Shen L, Liao K, Yang E, Yang F, Lin W, Wang J, Fan S, Huang X, Chen L, Shen H, Jin H, Ruan Y, Liu X, Zeng G, Xu JF, Pi J. Macrophage targeted iron oxide nanodecoys augment innate immunological and drug killings for more effective Mycobacterium Tuberculosis clearance. J Nanobiotechnology 2023; 21:369. [PMID: 37817142 PMCID: PMC10563239 DOI: 10.1186/s12951-023-02103-x] [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: 08/01/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb) infection, is still one of the top killers worldwide among infectious diseases. The escape of Mtb from immunological clearance and the low targeting effects of anti-TB drugs remain the substantial challenges for TB control. Iron is particularly required for Mtb growth but also toxic for Mtb in high dosages, which makes iron an ideal toxic decoy for the 'iron-tropic' Mtb. Here, a macrophage-targeted iron oxide nanoparticles (IONPs)-derived IONPs-PAA-PEG-MAN nanodecoy is designed to augment innate immunological and drug killings against intracellular Mtb. IONPs-PAA-PEG-MAN nanodecoy exhibits preferential uptake in macrophages to significantly increase drug uptake with sustained high drug contents in host cells. Moreover, it can serve as a specific nanodecoy for the 'iron-tropic' Mtb to realize the localization of Mtb contained phagosomes surrounding the drug encapsulated nanodecoys and co-localization of Mtb with the drug encapsulated nanodecoys in lysosomes, where the incorporated rifampicin (Rif) can be readily released under acidic lysosomal condition for enhanced Mtb killing. This drug encapsulated nanodecoy can also polarize Mtb infected macrophages into anti-mycobacterial M1 phenotype and enhance M1 macrophage associated pro-inflammatory cytokine (TNF-α) production to trigger innate immunological responses against Mtb. Collectively, Rif@IONPs-PAA-PEG-MAN nanodecoy can synergistically enhance the killing efficiency of intracellular Mtb in in vitro macrophages and ex vivo monocyte-derived macrophages, and also significantly reduce the mycobacterial burdens in the lung of infected mice with alleviated pathology. These results indicate that Rif@IONPs-PAA-PEG-MAN nanodecoy may have a potential for the development of more effective therapeutic strategy against TB by manipulating augmented innate immunity and drug killings.
Collapse
Affiliation(s)
- Ling Shen
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA.
| | - Kangsheng Liao
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Enzhuo Yang
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fen Yang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Wensen Lin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Jiajun Wang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Shuhao Fan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Xueqin Huang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Lingming Chen
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Jin
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Yongdui Ruan
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xing Liu
- Key Laboratory of Animal Disease Diagnostics and Immunology, Ministry of Agriculture, MOE International Joint Collaborative Research Laboratory for Animal Health & Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Gucheng Zeng
- Department of Microbiology, Zhongshan School of Medicine, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
| | - Jiang Pi
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China.
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, The Marine Biomedical Research Institute of Guangdong Medical University, ZhanJiang, Guangdong, China.
- Institute of Laboratory Medicine, School of Medical Technology, Guangdong Medical University, Dongguan, China.
| |
Collapse
|
8
|
Marzaman ANF, Roska TP, Sartini S, Utami RN, Sulistiawati S, Enggi CK, Manggau MA, Rahman L, Shastri VP, Permana AD. Recent Advances in Pharmaceutical Approaches of Antimicrobial Agents for Selective Delivery in Various Administration Routes. Antibiotics (Basel) 2023; 12:822. [PMID: 37237725 PMCID: PMC10215767 DOI: 10.3390/antibiotics12050822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Globally, the increase of pathogenic bacteria with antibiotic-resistant characteristics has become a critical challenge in medical treatment. The misuse of conventional antibiotics to treat an infectious disease often results in increased resistance and a scarcity of effective antimicrobials to be used in the future against the organisms. Here, we discuss the rise of antimicrobial resistance (AMR) and the need to combat it through the discovery of new synthetic or naturally occurring antibacterial compounds, as well as insights into the application of various drug delivery approaches delivered via various routes compared to conventional delivery systems. AMR-related infectious diseases are also discussed, as is the efficiency of various delivery systems. Future considerations in developing highly effective antimicrobial delivery devices to address antibiotic resistance are also presented here, especially on the smart delivery system of antibiotics.
Collapse
Affiliation(s)
- Ardiyah Nurul Fitri Marzaman
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Tri Puspita Roska
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Sartini Sartini
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Rifka Nurul Utami
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Sulistiawati Sulistiawati
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Cindy Kristina Enggi
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Marianti A. Manggau
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Latifah Rahman
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| | - Venkatram Prasad Shastri
- Institute for Macromolecular Chemistry, Albert Ludwigs Universitat Freiburg, 79085 Freiburg, Germany;
| | - Andi Dian Permana
- Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia; (A.N.F.M.); (T.P.R.); (S.S.); (R.N.U.); (S.S.); (C.K.E.); (M.A.M.); (L.R.)
| |
Collapse
|
9
|
Chopra H, Mohanta YK, Rauta PR, Ahmed R, Mahanta S, Mishra PK, Panda P, Rabaan AA, Alshehri AA, Othman B, Alshahrani MA, Alqahtani AS, AL Basha BA, Dhama K. An Insight into Advances in Developing Nanotechnology Based Therapeutics, Drug Delivery, Diagnostics and Vaccines: Multidimensional Applications in Tuberculosis Disease Management. Pharmaceuticals (Basel) 2023; 16:581. [PMID: 37111338 PMCID: PMC10145450 DOI: 10.3390/ph16040581] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/29/2023] Open
Abstract
Tuberculosis (TB), one of the deadliest contagious diseases, is a major concern worldwide. Long-term treatment, a high pill burden, limited compliance, and strict administration schedules are all variables that contribute to the development of MDR and XDR tuberculosis patients. The rise of multidrug-resistant strains and a scarcity of anti-TB medications pose a threat to TB control in the future. As a result, a strong and effective system is required to overcome technological limitations and improve the efficacy of therapeutic medications, which is still a huge problem for pharmacological technology. Nanotechnology offers an interesting opportunity for accurate identification of mycobacterial strains and improved medication treatment possibilities for tuberculosis. Nano medicine in tuberculosis is an emerging research field that provides the possibility of efficient medication delivery using nanoparticles and a decrease in drug dosages and adverse effects to boost patient compliance with therapy and recovery. Due to their fascinating characteristics, this strategy is useful in overcoming the abnormalities associated with traditional therapy and leads to some optimization of the therapeutic impact. It also decreases the dosing frequency and eliminates the problem of low compliance. To develop modern diagnosis techniques, upgraded treatment, and possible prevention of tuberculosis, the nanoparticle-based tests have demonstrated considerable advances. The literature search was conducted using Scopus, PubMed, Google Scholar, and Elsevier databases only. This article examines the possibility of employing nanotechnology for TB diagnosis, nanotechnology-based medicine delivery systems, and prevention for the successful elimination of TB illnesses.
Collapse
Affiliation(s)
- Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Yugal Kishore Mohanta
- Nanobiotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), Techno City, 9th Mile, Ri-Bhoi, Baridua 793101, Meghalaya, India
| | | | - Ramzan Ahmed
- Nanobiotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya (USTM), Techno City, 9th Mile, Ri-Bhoi, Baridua 793101, Meghalaya, India
- Department of Physics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Saurov Mahanta
- National Institute of Electronics and Information Technology (NIELIT), Guwahati Centre, Guwahati 781008, Assam, India
| | | | - Paramjot Panda
- School of Biological Sciences, AIPH University, Bhubaneswar 754001, Odisha, India
| | - Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Ahmad A. Alshehri
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Basim Othman
- Department of Public Health, Faculty of Applied Medical Sciences, Albaha University, Albaha 65779, Saudi Arabia
| | - Mohammed Abdulrahman Alshahrani
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 61441, Saudi Arabia
| | - Ali S. Alqahtani
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Khalid University, Abha 61481, Saudi Arabia
| | - Baneen Ali AL Basha
- Laboratory Department, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| |
Collapse
|
10
|
Inclusion complexation and liposomal encapsulation of an isoniazid hydrazone derivative in cyclodextrin for pH-dependent controlled release. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
11
|
Nanosized Drug Delivery Systems to Fight Tuberculosis. Pharmaceutics 2023; 15:pharmaceutics15020393. [PMID: 36839715 PMCID: PMC9964171 DOI: 10.3390/pharmaceutics15020393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Tuberculosis (TB) is currently the second deadliest infectious disease. Existing antitubercular therapies are long, complex, and have severe side effects that result in low patient compliance. In this context, nanosized drug delivery systems (DDSs) have the potential to optimize the treatment's efficiency while reducing its toxicity. Hundreds of publications illustrate the growing interest in this field. In this review, the main challenges related to the use of drug nanocarriers to fight TB are overviewed. Relevant publications regarding DDSs for the treatment of TB are classified according to the encapsulated drugs, from first-line to second-line drugs. The physicochemical and biological properties of the investigated formulations are listed. DDSs could simultaneously (i) optimize the therapy's antibacterial effects; (ii) reduce the doses; (iii) reduce the posology; (iv) diminish the toxicity; and as a global result, (v) mitigate the emergence of resistant strains. Moreover, we highlight that host-directed therapy using nanoparticles (NPs) is a recent promising trend. Although the research on nanosized DDSs for TB treatment is expanding, clinical applications have yet to be developed. Most studies are only dedicated to the development of new formulations, without the in vivo proof of concept. In the near future, it is expected that NPs prepared by "green" scalable methods, with intrinsic antibacterial properties and capable of co-encapsulating synergistic drugs, may find applications to fight TB.
Collapse
|
12
|
Liu F, Anton N, Niko Y, Klymchenko AS. Controlled Release and Capture of Aldehydes by Dynamic Imine Chemistry in Nanoemulsions: From Delivery to Detoxification. ACS APPLIED BIO MATERIALS 2023; 6:246-256. [PMID: 36516427 DOI: 10.1021/acsabm.2c00861] [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: 12/15/2022]
Abstract
Current biomedical applications of nanocarriers are focused on drug delivery, where encapsulated cargo is released in the target tissues under the control of external stimuli. Here, we propose a very different approach, where the active toxic molecules are removed from biological tissues by the nanocarrier. It is based on the drug-sponge concept, where specific molecules are captured by the lipid nanoemulsion (NE) droplets due to dynamic covalent chemistry inside their oil core. To this end, we designed a highly lipophilic amine (LipoAmine) capable of reacting with a free cargo-aldehyde (fluorescent dye and 4-hydroxynonenal toxin) directly inside lipid NEs, yielding a lipophilic imine conjugate well encapsulated in the oil core. The formation of imine bonds was first validated using a push-pull pyrene aldehyde dye, which changes its emission color during the reaction. The conjugate formation was independently confirmed by mass spectrometry. As a result, LipoAmine-loaded NEs spontaneously loaded cargo-aldehydes, yielding formulations stable against leakage at pH 7.4, which can further release the cargo in a low pH range (4-6) in solutions and living cells. Using fluorescence microscopy, we showed that LipoAmine NEs can extract pyrene aldehyde dye from cells as well as from an epithelial tissue (chicken skin). Moreover, successful extraction from cells was also achieved for a highly toxic aliphatic aldehyde 4-hydroxynonenal, which allowed obtaining the proof of concept for detoxification of living cells. Taken together, these results show that the dynamic imine chemistry inside NEs can be used to develop detoxification platforms.
Collapse
Affiliation(s)
- Fei Liu
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 route du Rhin, Illkirch 67401, France.,INSERM UMR 1260, Regenerative Nanomedicine (RNM), CRBS, Université de Strasbourg, Strasbourg 67000, France
| | - Nicolas Anton
- INSERM UMR 1260, Regenerative Nanomedicine (RNM), CRBS, Université de Strasbourg, Strasbourg 67000, France
| | - Yosuke Niko
- Research and Education Faculty, Multidisciplinary Science Cluster, Interdisciplinary Science Unit, Kochi University, 2-5-1, Akebono-cho, Kochi-shi, Kochi 780-8520, Japan
| | - Andrey S Klymchenko
- Laboratoire de Bioimagerie et Pathologies, UMR 7021 CNRS, Université de Strasbourg, 74 route du Rhin, Illkirch 67401, France
| |
Collapse
|
13
|
Feng W, Chittò M, Moriarty TF, Li G, Wang X. Targeted Drug Delivery Systems for Eliminating Intracellular Bacteria. Macromol Biosci 2023; 23:e2200311. [PMID: 36189899 DOI: 10.1002/mabi.202200311] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Indexed: 01/19/2023]
Abstract
The intracellular survival of pathogenic bacteria requires a range of survival strategies and virulence factors. These infections are a significant clinical challenge, wherein treatment frequently fails because of poor antibiotic penetration, stability, and retention in host cells. Drug delivery systems (DDSs) are promising tools to overcome these shortcomings and enhance the efficacy of antibiotic therapy. In this review, the classification and the mechanisms of intracellular bacterial persistence are elaborated. Furthermore, the systematic design strategies applied to DDSs to eliminate intracellular bacteria are also described, and the strategies used for internalization, intracellular activation, bacterial targeting, and immune enhancement are highlighted. Finally, this overview provides guidance for constructing functionalized DDSs to effectively eliminate intracellular bacteria.
Collapse
Affiliation(s)
- Wenli Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.,AO Research Institute Davos, Davos, 7270, Switzerland
| | - Marco Chittò
- AO Research Institute Davos, Davos, 7270, Switzerland
| | | | - Guofeng Li
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, P. R. China
| |
Collapse
|
14
|
Ammerman NC, Nuermberger EL, Owen A, Rannard SP, Meyers CF, Swindells S. Potential Impact of Long-Acting Products on the Control of Tuberculosis: Preclinical Advancements and Translational Tools in Preventive Treatment. Clin Infect Dis 2022; 75:S510-S516. [PMID: 36410384 PMCID: PMC10200320 DOI: 10.1093/cid/ciac672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A key component of global tuberculosis (TB) control is the treatment of latent TB infection. The use of long-acting technologies to administer TB preventive treatment has the potential to significantly improve the delivery and impact of this important public health intervention. For example, an ideal long-acting treatment could consist of a single dose that could be administered in the clinic (ie, a "1-shot cure" for latent TB). Interest in long-acting formulations for TB preventive therapy has gained considerable traction in recent years. This article presents an overview of the specific considerations and current preclinical advancements relevant for the development of long-acting technologies of TB drugs for treatment of latent infection, including attributes of target product profiles, suitability of drugs for long-acting formulations, ongoing research efforts, and translation to clinical studies.
Collapse
Affiliation(s)
- Nicole C Ammerman
- Department of Medical Microbiology and Infectious Diseases, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
- Center for Tuberculosis Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric L Nuermberger
- Center for Tuberculosis Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Owen
- Centre of Excellence for Long-acting Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Steve P Rannard
- Centre of Excellence for Long-acting Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Caren Freel Meyers
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Susan Swindells
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
15
|
Yang Z, Lou C, Wang X, Wang C, Shi Z, Niu N. Preparation, characterization, and in-vitro cytotoxicity of nanoliposomes loaded with anti-tubercular drugs and TGF-β1 siRNA for improving spinal tuberculosis therapy. BMC Infect Dis 2022; 22:824. [PMID: 36348467 PMCID: PMC9644586 DOI: 10.1186/s12879-022-07791-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
Background Tuberculosis (TB) represents a bacterial infection affecting many individuals each year and potentially leading to death. Overexpression of transforming growth factor (TGF)-β1 has a primary immunomodulatory function in human tuberculosis. This work aimed to develop nanoliposomes to facilitate the delivery of anti-tubercular products to THP-1-derived human macrophages as Mycobacterium host cells and to evaluate drug efficiencies as well as the effects of a TGF-β1-specific short interfering RNA (siRNA) delivery system employing nanoliposomes.
Methods In the current study, siTGF-β1 nanoliposomes loaded with the anti-TB drugs HRZ (isoniazid, rifampicin, and pyrazinamide) were prepared and characterized in vitro, determining the size, zeta potential, morphology, drug encapsulation efficiency (EE), cytotoxicity, and gene silencing efficiency of TGF-β1 siRNA.
Results HRZ/siTGF-β1 nanoliposomes appeared as smooth spheres showing the size and positive zeta potential of 168.135 ± 0.5444 nm and + 4.03 ± 1.32 mV, respectively. Drug EEs were 90%, 88%, and 37% for INH, RIF, and PZA, respectively. Meanwhile, the nanoliposomes were weakly cytotoxic towards human macrophages as assessed by the MTT assay. Nanoliposomal siTGF-β1 could significantly downregulate TGF-β1 in THP-1-derived human macrophages in vitro. Conclusion These findings suggested that HRZ-loaded nanoliposomes with siTGF-β1 have the potential for improving spinal tuberculosis chemotherapy via nano-encapsulation of anti-TB drugs.
Collapse
|
16
|
Vallet-Regí M, Schüth F, Lozano D, Colilla M, Manzano M. Engineering mesoporous silica nanoparticles for drug delivery: where are we after two decades? Chem Soc Rev 2022; 51:5365-5451. [PMID: 35642539 PMCID: PMC9252171 DOI: 10.1039/d1cs00659b] [Citation(s) in RCA: 121] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The present review details a chronological description of the events that took place during the development of mesoporous materials, their different synthetic routes and their use as drug delivery systems. The outstanding textural properties of these materials quickly inspired their translation to the nanoscale dimension leading to mesoporous silica nanoparticles (MSNs). The different aspects of introducing pharmaceutical agents into the pores of these nanocarriers, together with their possible biodistribution and clearance routes, would be described here. The development of smart nanocarriers that are able to release a high local concentration of the therapeutic cargo on-demand after the application of certain stimuli would be reviewed here, together with their ability to deliver the therapeutic cargo to precise locations in the body. The huge progress in the design and development of MSNs for biomedical applications, including the potential treatment of different diseases, during the last 20 years will be collated here, together with the required work that still needs to be done to achieve the clinical translation of these materials. This review was conceived to stand out from past reports since it aims to tell the story of the development of mesoporous materials and their use as drug delivery systems by some of the story makers, who could be considered to be among the pioneers in this area.
Collapse
Affiliation(s)
- María Vallet-Regí
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Ferdi Schüth
- Department of Heterogeneous Catalysis, Max-Planck-Institut für Kohlenforschung, Kaiser-Wilhelm-Platz 1, D-45470 Mülheim an der Ruhr, Germany
| | - Daniel Lozano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Montserrat Colilla
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| | - Miguel Manzano
- Chemistry in Pharmaceutical Sciences, School of Pharmacy, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i + 12), Pz/Ramón y Cajal s/n, Madrid 28040, Spain.
- Networking Research Centre on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid 28029, Spain
| |
Collapse
|
17
|
Forest V, Pourchez J. Nano-delivery to the lung - by inhalation or other routes and why nano when micro is largely sufficient? Adv Drug Deliv Rev 2022; 183:114173. [PMID: 35217112 DOI: 10.1016/j.addr.2022.114173] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/12/2022] [Accepted: 02/17/2022] [Indexed: 12/25/2022]
Abstract
Respiratory diseases gather a wide range of disorders which are generally difficult to treat, partly due to a poor delivery of drugs to the lung with adequate dose and minimum side effects. With the recent developments of nanotechnology, nano-delivery systems have raised interest. In this review, we detail the main types of nanocarriers that have been developed presenting their respective advantages and limitations. We also discuss the route of administration (systemic versus by inhalation), also considering technical aspects (different types of aerosol devices) with concrete examples of applications. Finally, we propose some perspectives of development in the field such as the nano-in-micro approaches, the emergence of drug vaping to generate airborne carriers in the submicron size range, the development of innovative respiratory models to assess regional aerosol deposition of nanoparticles or the application of nano-delivery to the lung in the treatment of other diseases.
Collapse
|
18
|
Wang W, Guo H, Lin S, Xiao X, Liu Y, Wang Y, Zhou D. Biosafety materials for tuberculosis treatment. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
19
|
Sarkar K, Kumar M, Jha A, Bharti K, Das M, Mishra B. Nanocarriers for tuberculosis therapy: Design of safe and effective drug delivery strategies to overcome the therapeutic challenges. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.102850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
20
|
Álvarez E, González B, Lozano D, Doadrio AL, Colilla M, Izquierdo-Barba I. Nanoantibiotics Based in Mesoporous Silica Nanoparticles: New Formulations for Bacterial Infection Treatment. Pharmaceutics 2021; 13:2033. [PMID: 34959315 PMCID: PMC8703556 DOI: 10.3390/pharmaceutics13122033] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/16/2022] Open
Abstract
This review focuses on the design of mesoporous silica nanoparticles for infection treatment. Written within a general context of contributions in the field, this manuscript highlights the major scientific achievements accomplished by professor Vallet-Regí's research group in the field of silica-based mesoporous materials for drug delivery. The aim is to bring out her pivotal role on the envisage of a new era of nanoantibiotics by using a deep knowledge on mesoporous materials as drug delivery systems and by applying cutting-edge technologies to design and engineer advanced nanoweapons to fight infection. This review has been divided in two main sections: the first part overviews the influence of the textural and chemical properties of silica-based mesoporous materials on the loading and release of antibiotic molecules, depending on the host-guest interactions. Furthermore, this section also remarks on the potential of molecular modelling in the design and comprehension of the performance of these release systems. The second part describes the more recent advances in the use of mesoporous silica nanoparticles as versatile nanoplatforms for the development of novel targeted and stimuli-responsive antimicrobial nanoformulations for future application in personalized infection therapies.
Collapse
Affiliation(s)
- Elena Álvarez
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (E.Á.); (B.G.); (D.L.); (A.L.D.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28040 Madrid, Spain
| | - Blanca González
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (E.Á.); (B.G.); (D.L.); (A.L.D.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28040 Madrid, Spain
| | - Daniel Lozano
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (E.Á.); (B.G.); (D.L.); (A.L.D.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28040 Madrid, Spain
| | - Antonio L. Doadrio
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (E.Á.); (B.G.); (D.L.); (A.L.D.)
| | - Montserrat Colilla
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (E.Á.); (B.G.); (D.L.); (A.L.D.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28040 Madrid, Spain
| | - Isabel Izquierdo-Barba
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria, Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; (E.Á.); (B.G.); (D.L.); (A.L.D.)
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28040 Madrid, Spain
| |
Collapse
|
21
|
García-Fernández A, Sancenón F, Martínez-Máñez R. Mesoporous silica nanoparticles for pulmonary drug delivery. Adv Drug Deliv Rev 2021; 177:113953. [PMID: 34474094 DOI: 10.1016/j.addr.2021.113953] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/11/2022]
Abstract
Over the last years, respiratory diseases represent a clinical concern, being included among the leading causes of death in the world due to the lack of effective lung therapies, mainly ascribed to the pulmonary barriers affecting the delivery of drugs to the lungs. In this way, nanomedicine has arisen as a promising approach to overcome the limitations of current therapies for pulmonary diseases. The use of nanoparticles allows enhancing drug bioavailability at the target site while minimizing undesired side effects. Despite different approaches have been developed for pulmonary delivery of drugs, including the use of polymers, lipid-based nanoparticles, and inorganic nanoparticles, more efforts are required to achieve effective pulmonary drug delivery. This review provides an overview of the clinical challenges in main lung diseases, as well as highlighted the role of nanomedicine in achieving efficient pulmonary drug delivery. Drug delivery into the lungs is a complex process limited by the anatomical, physiological and immunological barriers of the respiratory system. We discuss how nanomedicine can be useful to overcome these pulmonary barriers and give insights for the rational design of future nanoparticles for enhancing lung treatments. We also attempt herein to display more in detail the potential of mesoporous silica nanoparticles (MSNs) as promising nanocarrier for pulmonary drug delivery by providing a comprehensive overview of their application in lung delivery to date while discussing the use of these particles for the treatment of respiratory diseases.
Collapse
Affiliation(s)
- Alba García-Fernández
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Spain, Camino de Vera s/n, 46022 València, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Valencia, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 València, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Félix Sancenón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Spain, Camino de Vera s/n, 46022 València, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Valencia, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 València, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Ramón Martínez-Máñez
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Spain, Camino de Vera s/n, 46022 València, Spain; Unidad Mixta UPV-CIPF de Investigación en Mecanismos de Enfermedades y Nanomedicina, Valencia, Universitat Politècnica de València, Centro de Investigación Príncipe Felipe, 46012 València, Spain; Unidad Mixta de Investigación en Nanomedicina y Sensores. Universitat Politècnica de València, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
22
|
Lunn AM, Unnikrishnan M, Perrier S. Dual pH-Responsive Macrophage-Targeted Isoniazid Glycoparticles for Intracellular Tuberculosis Therapy. Biomacromolecules 2021; 22:3756-3768. [PMID: 34339606 DOI: 10.1021/acs.biomac.1c00554] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Tuberculosis (TB) is a global epidemic that kills over a million people every year, particularly in low-resource communities. Mycobacterium tuberculosis, the most common bacterium that causes TB, is difficult to treat, particularly in its latent phase, in part due to its ability to survive and replicate within the host macrophage. New therapeutic approaches resulting in better tolerated and shorter antibiotic courses that target intracellular bacteria are critical to effective treatment. The development of a novel, pH-responsive, mannosylated nanoparticle, covalently linked with isoniazid, a first-line TB antibiotic, is presented. This nanoparticle drug delivery agent has increased macrophage uptake and, upon exposure to the acidic phagolysosome, releases isoniazid through hydrolysis of a hydrazone bond, and disintegrates into a linear polymer. Full antibiotic activity is shown to be retained, with mannosylated isoniazid particles being the only treatment exhibiting complete bacterial eradication of intracellular bacteria, compared to an equivalent PEGylated system and free isoniazid. Such a system, able to effectively kill intracellular mycobacteria, holds promise for improved outcomes in TB infection.
Collapse
Affiliation(s)
- Andrew M Lunn
- Department of Chemistry, The University of Warwick, Gibbet Hill, Coventry CV4 7AL, U.K
| | - Meera Unnikrishnan
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, U.K
| | - Sébastien Perrier
- Department of Chemistry, The University of Warwick, Gibbet Hill, Coventry CV4 7AL, U.K.,Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
23
|
Mazlan MKN, Mohd Tazizi MHD, Ahmad R, Noh MAA, Bakhtiar A, Wahab HA, Mohd Gazzali A. Antituberculosis Targeted Drug Delivery as a Potential Future Treatment Approach. Antibiotics (Basel) 2021; 10:antibiotics10080908. [PMID: 34438958 PMCID: PMC8388690 DOI: 10.3390/antibiotics10080908] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 01/17/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the microorganism that causes tuberculosis. This infectious disease has been around for centuries, with the earliest record of Mtb around three million years ago. The discovery of the antituberculosis agents in the 20th century has managed to improve the recovery rate and reduce the death rate tremendously. However, the conventional antituberculosis therapy is complicated by the development of resistant strains and adverse drug reactions experienced by the patients. Research has been conducted continuously to discover new, safe, and effective antituberculosis drugs. In the last 50 years, only two molecules were approved despite laborious work and costly research. The repurposing of drugs is also being done with few drugs; antibiotics, particularly, were found to have antituberculosis activity. Besides the discovery work, enhancing the delivery of currently available antituberculosis drugs is also being researched. Targeted drug delivery may be a potentially useful approach to be developed into clinically accepted treatment modalities. Active targeting utilizes a specifically designed targeting agent to deliver a chemically conjugated drug(s) towards Mtb. Passive targeting is very widely explored, with the development of multiple types of nanoparticles from organic and inorganic materials. The nanoparticles will be engulfed by macrophages and this will eliminate the Mtb that is present in the macrophages, or the encapsulated drug may be released at the sites of infections that may be in the form of intra- and extrapulmonary tuberculosis. This article provided an overview on the history of tuberculosis and the currently available treatment options, followed by discussions on the discovery of new antituberculosis drugs and active and passive targeting approaches against Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Mohd Khairul Nizam Mazlan
- CHEST, School of Pharmaceutical Sciences, Sains@USM, Universiti Sains Malaysia, Bayan Lepas 11900, Malaysia; (M.K.N.M.); (R.A.)
| | | | - Rosliza Ahmad
- CHEST, School of Pharmaceutical Sciences, Sains@USM, Universiti Sains Malaysia, Bayan Lepas 11900, Malaysia; (M.K.N.M.); (R.A.)
| | - Muhammad Amirul Asyraf Noh
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Malaysia; (M.H.D.M.T.); (M.A.A.N.)
| | - Athirah Bakhtiar
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Malaysia;
| | - Habibah A. Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Malaysia; (M.H.D.M.T.); (M.A.A.N.)
- Correspondence: (H.A.W.); (A.M.G.)
| | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor 11800, Malaysia; (M.H.D.M.T.); (M.A.A.N.)
- Correspondence: (H.A.W.); (A.M.G.)
| |
Collapse
|
24
|
Beitzinger B, Gerbl F, Vomhof T, Schmid R, Noschka R, Rodriguez A, Wiese S, Weidinger G, Ständker L, Walther P, Michaelis J, Lindén M, Stenger S. Delivery by Dendritic Mesoporous Silica Nanoparticles Enhances the Antimicrobial Activity of a Napsin-Derived Peptide Against Intracellular Mycobacterium tuberculosis. Adv Healthc Mater 2021; 10:e2100453. [PMID: 34142469 PMCID: PMC11468746 DOI: 10.1002/adhm.202100453] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/20/2021] [Indexed: 12/28/2022]
Abstract
Tuberculosis remains a serious global health problem causing 1.3 million deaths annually. The causative pathogen Mycobacterium tuberculosis (Mtb) has developed several mechanisms to evade the immune system and resistances to many conventional antibiotics, so that alternative treatment strategies are urgently needed. By isolation from bronchoalveolar lavage and peptide optimization, a new antimicrobial peptide named NapFab is discovered. While showing robust activity against extracellular Mtb, the activity of NapFab against intracellular bacteria is limited due to low intracellular availability. By loading NapFab onto dendritic mesoporous silica nanoparticles (DMSN) as a carrier system, cellular uptake, and consequently antimycobacterial activity against intracellular Mtb is significantly enhanced. Furthermore, using lattice light-sheet fluorescence microscopy, it can be shown that the peptide is gradually released from the DMSN inside living macrophages over time. By electron microscopy and tomography, it is demonstrated that peptide loaded DMSN are stored in vesicular structures in proximity to mycobacterial phagosomes inside the cells, but the nanoparticles are typically not in direct contact with the bacteria. Based on the combination of functional and live-cell imaging analyses, it is hypothesized that after being released from the DMSN NapFab is able to enter the bacterial phagosome and gain access to the bacilli.
Collapse
Affiliation(s)
- Bastian Beitzinger
- Institute of Inorganic Chemistry IIUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Fabian Gerbl
- Institute of Medical Microbiology and HygieneUlm University HospitalAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Thomas Vomhof
- Institute of BiophysicsUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Roman Schmid
- Institute of Inorganic Chemistry IIUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Reiner Noschka
- Institute of Medical Microbiology and HygieneUlm University HospitalAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Armando Rodriguez
- Core Facility of Functional PeptidomicsUlm UniversityMeyerhofstraße 4Ulm89081Germany
- Core Unit of Mass Spectrometry and ProteomicsUlm UniversityAlbert‐Einstein Allee 11Ulm89081Germany
| | - Sebastian Wiese
- Core Unit of Mass Spectrometry and ProteomicsUlm UniversityAlbert‐Einstein Allee 11Ulm89081Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular BiologyUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Ludger Ständker
- Core Facility of Functional PeptidomicsUlm UniversityMeyerhofstraße 4Ulm89081Germany
| | - Paul Walther
- Central Facility for Electron MicroscopyUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Jens Michaelis
- Institute of BiophysicsUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Mika Lindén
- Institute of Inorganic Chemistry IIUlm UniversityAlbert‐Einstein‐Allee 11Ulm89081Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and HygieneUlm University HospitalAlbert‐Einstein‐Allee 11Ulm89081Germany
| |
Collapse
|
25
|
Zhong W, Zhang X, Zeng Y, Lin D, Wu J. Recent applications and strategies in nanotechnology for lung diseases. NANO RESEARCH 2021; 14:2067-2089. [PMID: 33456721 PMCID: PMC7796694 DOI: 10.1007/s12274-020-3180-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/11/2020] [Accepted: 10/11/2020] [Indexed: 05/14/2023]
Abstract
Lung diseases, including COVID-19 and lung cancers, is a huge threat to human health. However, for the treatment and diagnosis of various lung diseases, such as pneumonia, asthma, cancer, and pulmonary tuberculosis, are becoming increasingly challenging. Currently, several types of treatments and/or diagnostic methods are used to treat lung diseases; however, the occurrence of adverse reactions to chemotherapy, drug-resistant bacteria, side effects that can be significantly toxic, and poor drug delivery necessitates the development of more promising treatments. Nanotechnology, as an emerging technology, has been extensively studied in medicine. Several studies have shown that nano-delivery systems can significantly enhance the targeting of drug delivery. When compared to traditional delivery methods, several nanoparticle delivery strategies are used to improve the detection methods and drug treatment efficacy. Transporting nanoparticles to the lungs, loading appropriate therapeutic drugs, and the incorporation of intelligent functions to overcome various lung barriers have broad prospects as they can aid in locating target tissues and can enhance the therapeutic effect while minimizing systemic side effects. In addition, as a new and highly contagious respiratory infection disease, COVID-19 is spreading worldwide. However, there is no specific drug for COVID-19. Clinical trials are being conducted in several countries to develop antiviral drugs or vaccines. In recent years, nanotechnology has provided a feasible platform for improving the diagnosis and treatment of diseases, nanotechnology-based strategies may have broad prospects in the diagnosis and treatment of COVID-19. This article reviews the latest developments in nanotechnology drug delivery strategies in the lungs in recent years and studies the clinical application value of nanomedicine in the drug delivery strategy pertaining to the lung.
Collapse
Affiliation(s)
- Wenhao Zhong
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Xinyu Zhang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Yunxin Zeng
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Dongjun Lin
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
| | - Jun Wu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107 China
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006 China
| |
Collapse
|
26
|
Montalvo-Quirós S, Vallet-Regí M, Palacios A, Anguita J, Prados-Rosales RC, González B, Luque-Garcia JL. Mesoporous Silica Nanoparticles as a Potential Platform for Vaccine Development against Tuberculosis. Pharmaceutics 2020; 12:pharmaceutics12121218. [PMID: 33339306 PMCID: PMC7767215 DOI: 10.3390/pharmaceutics12121218] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/21/2023] Open
Abstract
The increasing emergence of new strains of Mycobacterium tuberculosis (Mtb) highly resistant to antibiotics constitute a public health issue, since tuberculosis still constitutes the primary cause of death in the world due to bacterial infection. Mtb has been shown to produce membrane-derived extracellular vesicles (EVs) containing proteins responsible for modulating the pathological immune response after infection. These natural vesicles were considered a promising alternative to the development of novel vaccines. However, their use was compromised by the observed lack of reproducibility between preparations. In this work, with the aim of developing nanosystems mimicking the extracellular vesicles produced by Mtb, mesoporous silica nanoparticles (MSNs) have been used as nanocarriers of immunomodulatory and vesicle-associated proteins (Ag85B, LprG and LprA). These novel nanosystems have been designed and extensively characterized, demonstrating the effectiveness of the covalent anchorage of the immunomodulatory proteins to the surface of the MSNs. The immunostimulatory capacity of the designed nanosystems has been demonstrated by measuring the levels of pro- (TNF) and anti-inflammatory (IL-10) cytokines in exposed macrophages. These results open a new possibility for the development of more complex nanosystems, including additional vesicle components or even antitubercular drugs, thus allowing for the combination of immunomodulatory and bactericidal effects against Mtb.
Collapse
Affiliation(s)
- Sandra Montalvo-Quirós
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain;
- Centro de Estudios Tecnológicos y Sociales y Facultad de Experimentales, Francisco de Vitoria University, 28223 Madrid, Spain
| | - María Vallet-Regí
- Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Complutense University of Madrid, 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Ainhoa Palacios
- Inflammation and Macrophage Plasticity Lab, CIC bioGUNE, 48160 Derio, Spain; (A.P.); (J.A.)
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Lab, CIC bioGUNE, 48160 Derio, Spain; (A.P.); (J.A.)
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Rafael C. Prados-Rosales
- Department of Preventive Medicine and Public Health and Microbiology, Faculty of Medicine, Autonomous University of Madrid, 28049 Madrid, Spain;
| | - Blanca González
- Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Complutense University of Madrid, 28040 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: (B.G.); (J.L.L.-G.)
| | - Jose L. Luque-Garcia
- Department of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, 28040 Madrid, Spain;
- Correspondence: (B.G.); (J.L.L.-G.)
| |
Collapse
|
27
|
Colilla M, Vallet-Regí M. Targeted Stimuli-Responsive Mesoporous Silica Nanoparticles for Bacterial Infection Treatment. Int J Mol Sci 2020; 21:E8605. [PMID: 33203098 PMCID: PMC7696808 DOI: 10.3390/ijms21228605] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023] Open
Abstract
The rise of antibiotic resistance and the growing number of biofilm-related infections make bacterial infections a serious threat for global human health. Nanomedicine has entered into this scenario by bringing new alternatives to design and develop effective antimicrobial nanoweapons to fight against bacterial infection. Among them, mesoporous silica nanoparticles (MSNs) exhibit unique characteristics that make them ideal nanocarriers to load, protect and transport antimicrobial cargoes to the target bacteria and/or biofilm, and release them in response to certain stimuli. The combination of infection-targeting and stimuli-responsive drug delivery capabilities aims to increase the specificity and efficacy of antimicrobial treatment and prevent undesirable side effects, becoming a ground-breaking alternative to conventional antibiotic treatments. This review focuses on the scientific advances developed to date in MSNs for infection-targeted stimuli-responsive antimicrobials delivery. The targeting strategies for specific recognition of bacteria are detailed. Moreover, the possibility of incorporating anti-biofilm agents with MSNs aimed at promoting biofilm penetrability is overviewed. Finally, a comprehensive description of the different scientific approaches for the design and development of smart MSNs able to release the antimicrobial payloads at the infection site in response to internal or external stimuli is provided.
Collapse
Affiliation(s)
- Montserrat Colilla
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28040 Madrid, Spain
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Unidad de Química Inorgánica y Bioinorgánica, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, 28040 Madrid, Spain
| |
Collapse
|
28
|
Dou Y, Li C, Li L, Guo J, Zhang J. Bioresponsive drug delivery systems for the treatment of inflammatory diseases. J Control Release 2020; 327:641-666. [PMID: 32911014 PMCID: PMC7476894 DOI: 10.1016/j.jconrel.2020.09.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Inflammation is intimately related to the pathogenesis of numerous acute and chronic diseases like cardiovascular disease, inflammatory bowel disease, rheumatoid arthritis, and neurodegenerative diseases. Therefore anti-inflammatory therapy is a very promising strategy for the prevention and treatment of these inflammatory diseases. To overcome the shortcomings of existing anti-inflammatory agents and their traditional formulations, such as nonspecific tissue distribution and uncontrolled drug release, bioresponsive drug delivery systems have received much attention in recent years. In this review, we first provide a brief introduction of the pathogenesis of inflammation, with an emphasis on representative inflammatory cells and mediators in inflammatory microenvironments that serve as pathological fundamentals for rational design of bioresponsive carriers. Then we discuss different materials and delivery systems responsive to inflammation-associated biochemical signals, such as pH, reactive oxygen species, and specific enzymes. Also, applications of various bioresponsive drug delivery systems in the treatment of typical acute and chronic inflammatory diseases are described. Finally, crucial challenges in the future development and clinical translation of bioresponsive anti-inflammatory drug delivery systems are highlighted.
Collapse
Affiliation(s)
- Yin Dou
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lanlan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Chemistry, College of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiawei Guo
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Pharmaceutical Analysis, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China; Institute of Burn Research, State Key Laboratory of Trauma, Burn and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
29
|
Montalvo-Quirós S, Gómez-Graña S, Vallet-Regí M, Prados-Rosales RC, González B, Luque-Garcia JL. Mesoporous silica nanoparticles containing silver as novel antimycobacterial agents against Mycobacterium tuberculosis. Colloids Surf B Biointerfaces 2020; 197:111405. [PMID: 33130523 DOI: 10.1016/j.colsurfb.2020.111405] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/14/2020] [Accepted: 10/04/2020] [Indexed: 12/31/2022]
Abstract
Tuberculosis remains today a major public health issue with a total of 9 million new cases and 2 million deaths annually. The lack of an effective vaccine and the increasing emergence of new strains of Mycobacterium tuberculosis (Mtb) highly resistant to antibiotics, anticipate a complicated scenario in the near future. The use of nanoparticles features as an alternative to antibiotics in tackling this problem due to their potential effectiveness in resistant bacterial strains. In this context, silver nanoparticles have demonstrated high bactericidal efficacy, although their use is limited by their relatively high toxicity, which calls for the design of nanocarriers that allow silver based nanoparticles to be safely delivered to the target cells or tissues. In this work mesoporous silica nanoparticles are used as carriers of silver based nanoparticles as antimycobacterial agent against Mtb. Two different synthetic approaches have been used to afford, on the one hand, a 2D hexagonal mesoporous silica nanosystem which contains silver bromide nanoparticles distributed all through the silica network and, on the other hand, a core@shell nanosystem with metallic silver nanoparticles as core and mesoporous silica shell in a radial mesoporous rearrangement. Both materials have demonstrated good antimycobacterial capacity in in vitro test using Mtb, being lower the minimum inhibitory concentration for the nanosystem which contains silver bromide. Therefore, the interaction of this material with the mycobacterial cell has been studied by cryo-electron microscopy, establishing a direct connection between the antimycobactericidal effect observed and the damage induced in the cell envelope.
Collapse
Affiliation(s)
- Sandra Montalvo-Quirós
- Departament of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Av. Complutense s/n, 28040, Madrid, Spain; Centro de Estudios Tecnológicos y Sociales y Facultad de Experimentales, Universidad Francisco de Vitoria, 28223, Pozuelo de Alarcón Madrid, Spain
| | - Sergio Gómez-Graña
- Departament of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - María Vallet-Regí
- Departament of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Plaza Ramón y Cajal s/n, 28040, Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Rafael C Prados-Rosales
- Department of Preventive Medicine and Public Health and Microbiology, Faculty of Medicine, Autonoma University of Madrid, Spain
| | - Blanca González
- Departament of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Plaza Ramón y Cajal s/n, 28040, Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Jose L Luque-Garcia
- Departament of Analytical Chemistry, Faculty of Chemistry, Complutense University of Madrid, Av. Complutense s/n, 28040, Madrid, Spain.
| |
Collapse
|
30
|
Carvalho GC, Sábio RM, de Cássia Ribeiro T, Monteiro AS, Pereira DV, Ribeiro SJL, Chorilli M. Highlights in Mesoporous Silica Nanoparticles as a Multifunctional Controlled Drug Delivery Nanoplatform for Infectious Diseases Treatment. Pharm Res 2020; 37:191. [PMID: 32895867 PMCID: PMC7476752 DOI: 10.1007/s11095-020-02917-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/25/2020] [Indexed: 12/19/2022]
Abstract
Infectious diseases are a major global concern being responsible for high morbidity and mortality mainly due to the development and enhancement of multidrug-resistant microorganisms exposing the fragility of medicines and vaccines commonly used to these treatments. Taking into account the scarcity of effective formulation to treat infectious diseases, nanotechnology offers a vast possibility of ground-breaking platforms to design new treatment through smart nanostructures for drug delivery purposes. Among the available nanosystems, mesoporous silica nanoparticles (MSNs) stand out due their multifunctionality, biocompatibility and tunable properties make them emerging and actual nanocarriers for specific and controlled drug release. Considering the high demand for diseases prevention and treatment, this review exploits the MSNs fabrication and their behavior in biological media besides highlighting the most of strategies to explore the wide MSNs functionality as engineered, smart and effective controlled drug release nanovehicles for infectious diseases treatment. Graphical Abstract Schematic representation of multifunctional MSNs-based nanoplatforms for infectious diseases treatment.
Collapse
Affiliation(s)
- Gabriela Corrêa Carvalho
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Rafael Miguel Sábio
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, 14800-903, Brazil.
| | - Tais de Cássia Ribeiro
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Andreia Sofia Monteiro
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara, 14800-060, Brazil
| | | | | | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| |
Collapse
|
31
|
Selvarajan V, Obuobi S, Ee PLR. Silica Nanoparticles-A Versatile Tool for the Treatment of Bacterial Infections. Front Chem 2020; 8:602. [PMID: 32760699 PMCID: PMC7374024 DOI: 10.3389/fchem.2020.00602] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/09/2020] [Indexed: 12/25/2022] Open
Abstract
The rapid emergence of drug resistance continues to outpace the development of new antibiotics in the treatment of infectious diseases. Conventional therapy is currently limited by drug access issues such as low intracellular drug accumulations, drug efflux by efflux pumps and/or enzymatic degradation. To improve access, targeted delivery using nanocarriers could provide the quantum leap in intracellular drug transport and retention. Silica nanoparticles (SiNPs) with crucial advantages such as large surface area, ease-of-functionalization, and biocompatibility, are one of the most commonly used nanoparticles in drug delivery applications. A porous variant, called the mesoporous silica nanoparticles (MSN), also confers additional amenities such as tunable pore size and volume, leading to high drug loading capacity. In the context of bacterial infections, SiNPs and its variants can act as a powerful tool for the targeted delivery of antimicrobials, potentially reducing the impact of high drug dosage and its side effects. In this review, we will provide an overview of SiNPs synthesis, its structural proficiency which is critical in loading and conjugation of antimicrobials and its role in different antimicrobial applications with emphasis on intracellular drug targeting in anti-tuberculosis therapy, nitric oxide delivery, and metal nanocomposites. The role of SiNPs in antibiofilm coatings will also be covered in the context of nosocomial infections and surgical implants.
Collapse
Affiliation(s)
- Vanitha Selvarajan
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Sybil Obuobi
- Drug Transport and Delivery Research Group, Department of Pharmacy, UIT The Arctic University of Norway, Tromsø, Norway
| | - Pui Lai Rachel Ee
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, Singapore, Singapore
| |
Collapse
|
32
|
Rodrigues B, Shende P. Monodispersed metal-based dendrimeric nanoclusters for potentiation of anti-tuberculosis action. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2020.112731] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
33
|
Pi J, Shen L, Yang E, Shen H, Huang D, Wang R, Hu C, Jin H, Cai H, Cai J, Zeng G, Chen ZW. Macrophage‐Targeted Isoniazid–Selenium Nanoparticles Promote Antimicrobial Immunity and Synergize Bactericidal Destruction of Tuberculosis Bacilli. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201912122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Jiang Pi
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
- Department of Microbiology Zhongshan School of Medicine Key Laboratory for Tropical Diseases Control of the Ministry of Education Sun Yat-sen University Guangzhou Guangdong 510080 China
| | - Ling Shen
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Enzhuo Yang
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis Shanghai Pulmonary Hospital Tongji University School of Medicine Shanghai 200433 China
| | - Dan Huang
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Richard Wang
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Chunmiao Hu
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Hua Jin
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| | - Huaihong Cai
- Department of Chemistry Jinan University Guangzhou Guangdong 510632 China
| | - Jiye Cai
- Department of Chemistry Jinan University Guangzhou Guangdong 510632 China
| | - Gucheng Zeng
- Department of Microbiology Zhongshan School of Medicine Key Laboratory for Tropical Diseases Control of the Ministry of Education Sun Yat-sen University Guangzhou Guangdong 510080 China
| | - Zheng W. Chen
- Department of Microbiology and Immunology Center for Primate Biomedical Research University of Illinois College of Medicine Chicago IL 60612 USA
| |
Collapse
|
34
|
Antitubercular nanocarrier monotherapy: Study of In Vivo efficacy and pharmacokinetics for rifampicin. J Control Release 2020; 321:312-323. [PMID: 32067995 DOI: 10.1016/j.jconrel.2020.02.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/07/2020] [Accepted: 02/13/2020] [Indexed: 02/08/2023]
Abstract
Tuberculosis represents a major global health problem for which improved approaches are needed to shorten the course of treatment and to combat the emergence of resistant strains. The development of effective and safe nanobead-based interventions can be particularly relevant for increasing the concentrations of antitubercular agents within the infected site and reducing the concentrations in the general circulation, thereby avoiding off-target toxic effects. In this work, rifampicin, a first-line antitubercular agent, was encapsulated into biocompatible and biodegradable polyester-based nanoparticles. In a well-established BALB/c mouse model of pulmonary tuberculosis, the nanoparticles provided improved pharmacokinetics and pharmacodynamics. The nanoparticles were well tolerated and much more efficient than an equivalent amount of free rifampicin.
Collapse
|
35
|
Pi J, Shen L, Yang E, Shen H, Huang D, Wang R, Hu C, Jin H, Cai H, Cai J, Zeng G, Chen ZW. Macrophage-Targeted Isoniazid-Selenium Nanoparticles Promote Antimicrobial Immunity and Synergize Bactericidal Destruction of Tuberculosis Bacilli. Angew Chem Int Ed Engl 2020; 59:3226-3234. [PMID: 31756258 DOI: 10.1002/anie.201912122] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Indexed: 12/16/2022]
Abstract
Pathogenesis hallmarks for tuberculosis (TB) are the Mycobacterium tuberculosis (Mtb) escape from phagolysosomal destruction and limited drug delivery into infected cells. Several nanomaterials can be entrapped in lysosomes, but the development of functional nanomaterials to promote phagolysosomal Mtb clearance remains a big challenge. Here, we report on the bactericidal effects of selenium nanoparticles (Se NPs) against Mtb and further introduce a novel nanomaterial-assisted anti-TB strategy manipulating Ison@Man-Se NPs for synergistic drug-induced and phagolysosomal destruction of Mtb. Ison@Man-Se NPs preferentially entered macrophages and accumulated in lysosomes releasing Isoniazid. Surprisingly, Ison@Man-Se/Man-Se NPs further promoted the fusion of Mtb into lysosomes for synergistic lysosomal and Isoniazid destruction of Mtb. Concurrently, Ison@Man-Se/Man-Se NPs also induced autophagy sequestration of Mtb, evolving into lysosome-associated autophagosomal Mtb degradation linked to ROS-mitochondrial and PI3K/Akt/mTOR signaling pathways. This novel nanomaterial-assisted anti-TB strategy manipulating antimicrobial immunity and Mtb clearance may potentially serve in more effective therapeutics against TB and drug-resistant TB.
Collapse
Affiliation(s)
- Jiang Pi
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA.,Department of Microbiology, Zhongshan School of Medicine, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Ling Shen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Enzhuo Yang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Hongbo Shen
- Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Dan Huang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Richard Wang
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Chunmiao Hu
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Hua Jin
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| | - Huaihong Cai
- Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Jiye Cai
- Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Gucheng Zeng
- Department of Microbiology, Zhongshan School of Medicine, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
| | - Zheng W Chen
- Department of Microbiology and Immunology, Center for Primate Biomedical Research, University of Illinois College of Medicine, Chicago, IL, 60612, USA
| |
Collapse
|
36
|
Jeon M, Lin G, Stephen ZR, Kato FL, Zhang M. Paclitaxel‐Loaded Iron Oxide Nanoparticles for Targeted Breast Cancer Therapy. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900081] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mike Jeon
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| | - Guanyou Lin
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| | - Zachary R. Stephen
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| | - Frances L. Kato
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| | - Miqin Zhang
- Department of Materials Science and Engineering University of Washington Seattle WA 98195 USA
| |
Collapse
|
37
|
Synthesis and Characterization of pH-Sensitive Inulin Conjugate of Isoniazid for Monocyte-Targeted Delivery. Pharmaceutics 2019; 11:pharmaceutics11110555. [PMID: 31661841 PMCID: PMC6920787 DOI: 10.3390/pharmaceutics11110555] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/14/2019] [Accepted: 10/25/2019] [Indexed: 12/13/2022] Open
Abstract
The use of particles for monocyte-mediated delivery could be a more efficient strategy and approach to achieve intracellular targeting and delivery of antitubercular drugs to host macrophages. In this study, the potential of inulin microparticles to serve as a drug vehicle in the treatment of chronic tuberculosis using a monocytes-mediated drug targeting approach was evaluated. Isoniazid (INH) was conjugated to inulin via hydrazone linkage in order to obtain a pH-sensitive inulin-INH conjugate. The conjugate was then characterized using proton nuclear magnetic resonance (1HNMR), Fourier transform infrared spectroscopy (FTIR) as well as in vitro, cellular uptake and intracellular Mycobacterium tuberculosis (Mtb) antibacterial efficacy. The acid-labile hydrazone linkage conferred pH sensitivity to the inulin-INH conjugate with ~95, 77 and 65% of the drug released after 5 h at pH 4.5, 5.2, and 6.0 respectively. Cellular uptake studies confirm that RAW 264.7 monocytic cells efficiently internalized the inulin conjugates into endocytic compartments through endocytosis. The intracellular efficacy studies demonstrate that the inulin conjugates possess a dose-dependent targeting effect against Mtb-infected monocytes. This was through efficient internalization and cleavage of the hydrazone bond by the acidic environment of the lysosome, which subsequently released the isoniazid intracellularly to the Mtb reservoir. These results clearly suggest that inulin conjugates can serve as a pH-sensitive intracellular drug delivery system for TB treatment.
Collapse
|
38
|
Nkanga CI, Krause RWM. Encapsulation of Isoniazid-conjugated Phthalocyanine-In-Cyclodextrin-In-Liposomes Using Heating Method. Sci Rep 2019; 9:11485. [PMID: 31391517 PMCID: PMC6685989 DOI: 10.1038/s41598-019-47991-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/26/2019] [Indexed: 12/27/2022] Open
Abstract
Liposomes are reputed colloidal vehicles that hold the promise for targeted delivery of anti-tubercular drugs (ATBDs) to alveolar macrophages that host Mycobacterium tuberculosis. However, the costly status of liposome technology, particularly due to the use of special manufacture equipment and expensive lipid materials, may preclude wider developments of therapeutic liposomes. In this study, we report efficient encapsulation of a complex system, consisting of isoniazid-hydrazone-phthalocyanine conjugate (Pc-INH) in gamma-cyclodextrin (γ-CD), in liposomes using crude soybean lecithin by means of a simple organic solvent-free method, heating method (HM). Inclusion complexation was performed in solution and solid-state, and evaluated using UV-Vis, magnetic circular dichroism, 1H NMR, diffusion ordered spectroscopy and FT-IR. The HM-liposomes afforded good encapsulation efficiency (71%) for such a large Pc-INH/γ-CD complex (PCD) system. The stability and properties of the PCD-HM-liposomes look encouraging; with particle size 240 nm and Zeta potential −57 mV that remained unchanged upon storage at 4 °C for 5 weeks. The release study performed in different pH media revealed controlled release profiles that went up to 100% at pH 4.4, from about 40% at pH 7.4. This makes PCD-liposomes a promising system for site-specific ATBD delivery, and a good example of simple liposomal encapsulation of large hydrophobic compounds.
Collapse
Affiliation(s)
- Christian Isalomboto Nkanga
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Rhodes University, PO Box 94, Grahamstown, 6140, Eastern Cape, South Africa
| | - Rui Werner Maçedo Krause
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Rhodes University, PO Box 94, Grahamstown, 6140, Eastern Cape, South Africa.
| |
Collapse
|
39
|
Roloff A, Nirmalananthan-Budau N, Rühle B, Borcherding H, Thiele T, Schedler U, Resch-Genger U. Quantification of Aldehydes on Polymeric Microbead Surfaces via Catch and Release of Reporter Chromophores. Anal Chem 2019; 91:8827-8834. [PMID: 31188569 DOI: 10.1021/acs.analchem.8b05515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aldehyde moieties on 2D-supports or micro- and nanoparticles can function as anchor groups for the attachment of biomolecules or as reversible binding sites for proteins on cell surfaces. The use of aldehyde-based materials in bioanalytical and medical settings calls for reliable methods to detect and quantify this functionality. We report here on a versatile concept to quantify the accessible aldehyde moieties on particle surfaces through the specific binding and subsequent release of small reporter molecules such as fluorescent dyes and nonfluorescent chromophores utilizing acylhydrazone formation as a reversible covalent labeling strategy. This is representatively demonstrated for a set of polymer microparticles with different aldehyde labeling densities. Excess reporter molecules can be easily removed by washing, eliminating inaccuracies caused by unspecific adsorption to hydrophobic surfaces. Cleavage of hydrazones at acidic pH assisted by a carbonyl trap releases the fluorescent reporters rapidly and quasi-quantitatively and allows for their fluorometric detection at low concentration. Importantly, this strategy separates the signal-generating molecules from the bead surface. This circumvents common issues associated with light scattering and signal distortions that are caused by binding-induced changes in reporter fluorescence as well as quenching dye-dye interactions on crowded particle surfaces. In addition, we demonstrate that the release of a nonfluorescent chromophore via disulfide cleavage and subsequent quantification by absorption spectroscopy gives comparable results, verifying that both assays are capable of rapid and sensitive quantification of aldehydes on microbead surfaces. These strategies enable a quantitative comparison of bead batches with different functionalization densities, and a qualitative prediction of their coupling efficiencies in bioconjugations, as demonstrated in reductive amination reactions with Streptavidin.
Collapse
Affiliation(s)
- Alexander Roloff
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany
| | - Nithiya Nirmalananthan-Budau
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany.,Institut für Chemie und Biochemie , Freie Universität Berlin , Takustrasse 3 , D-14195 Berlin , Germany
| | - Bastian Rühle
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany
| | | | - Thomas Thiele
- PolyAn GmbH , Rudolf-Baschant-Straße 2 , D-13086 Berlin , Germany
| | - Uwe Schedler
- PolyAn GmbH , Rudolf-Baschant-Straße 2 , D-13086 Berlin , Germany
| | - Ute Resch-Genger
- Federal Institute for Materials Research and Testing (BAM) , Richard-Willstätter-Straße 11 , D-12489 Berlin , Germany
| |
Collapse
|
40
|
Chen W, Glackin CA, Horwitz MA, Zink JI. Nanomachines and Other Caps on Mesoporous Silica Nanoparticles for Drug Delivery. Acc Chem Res 2019; 52:1531-1542. [PMID: 31082188 DOI: 10.1021/acs.accounts.9b00116] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesoporous silica nanoparticles (MSNs) are delivery vehicles that can carry cargo molecules and release them on command. The particles used in the applications reported in this Account are around 100 nm in diameter (about the size of a virus) and contain 2.5 nm tubular pores with a total volume of about 1 cm3/g. For the biomedical applications discussed here, the cargo is trapped in the pores until the particles are stimulated to release it. The challenges are to get the particles to the site of a disease and then to deliver the cargo on command. We describe methods to do both, and we illustrate the applicability of the particles to cure cancer and intracellular infectious disease. Our first steps were to design multifunctional nanoparticles with properties that allow them to carry and deliver hydrophobic drugs. Many important pharmaceuticals are hydrophobic and cannot reach the diseased sites by themselves. We describe how we modified MSNs to make them dispersible, imagable, and targetable and discuss in vitro studies. We then present examples of surface modifications that allow them to deliver large molecules such as siRNA. In vivo studies of siRNA delivery to treat triple-negative breast and ovarian cancers are presented. The next steps are to attach nanomachines and other types of caps that trap drug molecules but release them when stimulated. We describe nanomachines that respond autonomously (without human intervention) to stimuli specific to disease sites. A versatile type of machine is a nanovalve that is closed at neutral (blood) pH but opens upon acidification that occurs in endolysosomes of cancer cells. Another type of machine, a snap-top cap, is stimulated by reducing agents such as glutathione in the cytosol of cells. Both of these platforms were studied in vitro to deliver antibiotics to infected macrophages and in vivo to cure and kill the intracellular bacteria M. tuberculosis and F. tularensis. The latter is a tier 1 select agent of bioterrorism. Finally, we describe nanomachines for drug delivery that are controlled by externally administered light and magnetic fields. A futuristic dream for nanotherapy is the ability to control a nano-object everywhere in the body. Magnetic fields penetrate completely and have spatial selectivity governed by the size of the field-producing coil. We describe how to control nanovalves with alternating magnetic fields (AMFs) and superparamagnetic cores inside the MSNs. The AMF heats the cores, and temperature-sensitive caps release the cargo. In vitro studies demonstrate dose control of the therapeutic to cause apoptosis without overheating the cells. Nanocarriers have great promise for therapeutic applications, and MSNs that can carry drugs to the site of a disease to produce a high local concentration without premature release and off-target damage may have the capability of realizing this goal.
Collapse
Affiliation(s)
- Wei Chen
- Department of Chemistry & Biochemistry, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Carlotta A. Glackin
- Department of Stem Cell and Developmental Biology, City of Hope−Beckman Research Institute, Duarte, California 91010, United States
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, California 90095, United States
| | - Jeffrey I. Zink
- Department of Chemistry & Biochemistry, University of California, Los Angeles, California 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| |
Collapse
|
41
|
Trousil J, Syrová Z, Dal NJK, Rak D, Konefał R, Pavlova E, Matějková J, Cmarko D, Kubíčková P, Pavliš O, Urbánek T, Sedlák M, Fenaroli F, Raška I, Štěpánek P, Hrubý M. Rifampicin Nanoformulation Enhances Treatment of Tuberculosis in Zebrafish. Biomacromolecules 2019; 20:1798-1815. [PMID: 30785284 DOI: 10.1021/acs.biomac.9b00214] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mycobacterium tuberculosis, the etiologic agent of tuberculosis, is an intracellular pathogen of alveolar macrophages. These cells avidly take up nanoparticles, even without the use of specific targeting ligands, making the use of nanotherapeutics ideal for the treatment of such infections. Methoxy poly(ethylene oxide)- block-poly(ε-caprolactone) nanoparticles of several different polymer blocks' molecular weights and sizes (20-110 nm) were developed and critically compared as carriers for rifampicin, a cornerstone in tuberculosis therapy. The polymeric nanoparticles' uptake, consequent organelle targeting and intracellular degradation were shown to be highly dependent on the nanoparticles' physicochemical properties (the cell uptake half-lives 2.4-21 min, the degradation half-lives 51.6 min-ca. 20 h after the internalization). We show that the nanoparticles are efficiently taken up by macrophages and are able to effectively neutralize the persisting bacilli. Finally, we demonstrate, using a zebrafish model of tuberculosis, that the nanoparticles are well tolerated, have a curative effect, and are significantly more efficient compared to a free form of rifampicin. Hence, these findings demonstrate that this system shows great promise, both in vitro and in vivo, for the treatment of tuberculosis.
Collapse
Affiliation(s)
- Jiří Trousil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského náměstí 2 , 162 00 Prague 6 , Czech Republic.,Department of Analytical Chemistry, Faculty of Science , Charles University , Hlavova 8 , 128 43 Prague 2 , Czech Republic
| | - Zdeňka Syrová
- Institute of Biology and Medical Genetics, First Faculty of Medicine , Charles University and General University Hospital in Prague , Albertov 4 , 128 00 Prague 2 , Czech Republic
| | - Nils-Jørgen K Dal
- Department of Biosciences , University of Oslo , Blindernveien 31 , 0371 Oslo , Norway
| | - Dmytro Rak
- Institute of Experimental Physics , Slovak Academy of Sciences , Watsonova 47 , 040 01 Košice , Slovakia
| | - Rafał Konefał
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského náměstí 2 , 162 00 Prague 6 , Czech Republic
| | - Ewa Pavlova
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského náměstí 2 , 162 00 Prague 6 , Czech Republic
| | - Jana Matějková
- Department of Medical Microbiology, Second Faculty of Medicine , Charles University and Motol University Hospital , V Úvalu 84 , 150 06 Prague 5 , Czech Republic
| | - Dušan Cmarko
- Institute of Biology and Medical Genetics, First Faculty of Medicine , Charles University and General University Hospital in Prague , Albertov 4 , 128 00 Prague 2 , Czech Republic
| | - Pavla Kubíčková
- Center of Biological Defense , Military Health Institute, Military Medical Agency , 561 66 Těchonín , Czech Republic
| | - Oto Pavliš
- Center of Biological Defense , Military Health Institute, Military Medical Agency , 561 66 Těchonín , Czech Republic
| | - Tomáš Urbánek
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského náměstí 2 , 162 00 Prague 6 , Czech Republic
| | - Marián Sedlák
- Institute of Experimental Physics , Slovak Academy of Sciences , Watsonova 47 , 040 01 Košice , Slovakia
| | - Federico Fenaroli
- Department of Biosciences , University of Oslo , Blindernveien 31 , 0371 Oslo , Norway
| | - Ivan Raška
- Institute of Biology and Medical Genetics, First Faculty of Medicine , Charles University and General University Hospital in Prague , Albertov 4 , 128 00 Prague 2 , Czech Republic
| | - Petr Štěpánek
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského náměstí 2 , 162 00 Prague 6 , Czech Republic
| | - Martin Hrubý
- Institute of Macromolecular Chemistry, Czech Academy of Sciences , Heyrovského náměstí 2 , 162 00 Prague 6 , Czech Republic
| |
Collapse
|
42
|
Chen W, Cheng CA, Zink JI. Spatial, Temporal, and Dose Control of Drug Delivery using Noninvasive Magnetic Stimulation. ACS NANO 2019; 13:1292-1308. [PMID: 30633500 DOI: 10.1021/acsnano.8b06655] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Noninvasive stimuli-responsive drug delivery using magnetic fields in conjunction with superparamagnetic nanoparticles offers the potential for the spatial and temporal control of drug release. When hyperthermia is not desired and control of the dosage is required, it is necessary to design a platform in which local heating on the nanoscale releases the therapeutic cargo without the bulk heating of the surrounding medium. In this paper, we report a design using a stimuli-responsive nanoparticle platform to control the dosage of the cargo released by an alternating magnetic field (AMF) actuation. A core@shell structure with a superparamagnetic doped iron oxide (MnFe2O4@CoFe2O4) nanoparticle core in a mesoporous silica shell was synthesized. The core used here has a high saturation magnetization value and a high specific loss power for heat generation under an AMF. The mesoporous shell has a high cargo-carrying capacity. A thermoresponsive molecular-based gatekeeper containing an aliphatic azo group was modified on the core@shell nanoparticles to regulate the cargo release. The mesoporous structure of the silica shell remained intact after exposure to an AMF, showing that the release of cargo is due to the removal of the gatekeepers instead of the destruction of the structure. Most importantly, we demonstrated that the amount of cargo released could be adjusted by the AMF exposure time. By applying multiple sequential exposures of AMF, we were able to release the cargo step-wise and increase the total amount of released cargo. In vitro studies showed that the death of pancreatic cancer cells treated by drug-loaded nanoparticles was controlled by different lengths of AMF exposure time due to different amount of drugs released from the carriers. The strategy developed here holds great promise for achieving the dosage, temporal, and spatial control of therapeutics delivery without the risk of overheating the particles' surroundings.
Collapse
|
43
|
Clemens DL, Lee BY, Plamthottam S, Tullius MV, Wang R, Yu CJ, Li Z, Dillon BJ, Zink JI, Horwitz MA. Nanoparticle Formulation of Moxifloxacin and Intramuscular Route of Delivery Improve Antibiotic Pharmacokinetics and Treatment of Pneumonic Tularemia in a Mouse Model. ACS Infect Dis 2019; 5:281-291. [PMID: 30480992 DOI: 10.1021/acsinfecdis.8b00268] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Francisella tularensis causes a serious and often fatal infection, tularemia. We compared the efficacy of moxifloxacin formulated as free drug vs disulfide snap-top mesoporous silica nanoparticles (MSNs) in a mouse model of pneumonic tularemia. We found that MSN-formulated moxifloxacin was more effective than free drug and that the intramuscular and subcutaneous routes were markedly more effective than the intravenous route. Measurement of tissue silica levels and fluorescent flow cytometry assessment of colocalization of MSNs with infected cells revealed that the enhanced efficacy of MSNs and the intramuscular route of delivery was not due to better delivery of MSNs to infected tissues or cells. However, moxifloxacin blood levels demonstrated that the nanoparticle formulation and intramuscular route provided the longest half-life and longest time above the minimal inhibitory concentration. Thus, improved pharmacokinetics are responsible for the greater efficacy of nanoparticle formulation and intramuscular delivery compared with free drug and intravenous delivery.
Collapse
Affiliation(s)
- Daniel L. Clemens
- Division of Infectious Diseases, Department of Medicine, University of California, CHS 37-121, 10833 Le Conte Avenue, Los Angeles, California 90095-1688, United States
| | - Bai-Yu Lee
- Division of Infectious Diseases, Department of Medicine, University of California, CHS 37-121, 10833 Le Conte Avenue, Los Angeles, California 90095-1688, United States
| | - Sheba Plamthottam
- Department of Chemistry and Biochemistry, University of California, 3013 Young Drive East, Los Angeles, California 90095-1569, United States
| | - Michael V. Tullius
- Division of Infectious Diseases, Department of Medicine, University of California, CHS 37-121, 10833 Le Conte Avenue, Los Angeles, California 90095-1688, United States
| | - Ruining Wang
- Department of Chemistry and Biochemistry, University of California, 3013 Young Drive East, Los Angeles, California 90095-1569, United States
| | - Chia-Jung Yu
- Department of Chemistry and Biochemistry, University of California, 3013 Young Drive East, Los Angeles, California 90095-1569, United States
| | - Zilu Li
- Department of Chemistry and Biochemistry, University of California, 3013 Young Drive East, Los Angeles, California 90095-1569, United States
| | - Barbara Jane Dillon
- Division of Infectious Diseases, Department of Medicine, University of California, CHS 37-121, 10833 Le Conte Avenue, Los Angeles, California 90095-1688, United States
| | - Jeffrey I. Zink
- Department of Chemistry and Biochemistry, University of California, 3013 Young Drive East, Los Angeles, California 90095-1569, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095-8352, United States
| | - Marcus A. Horwitz
- Division of Infectious Diseases, Department of Medicine, University of California, CHS 37-121, 10833 Le Conte Avenue, Los Angeles, California 90095-1688, United States
| |
Collapse
|
44
|
Zhao N, Yan L, Zhao X, Chen X, Li A, Zheng D, Zhou X, Dai X, Xu FJ. Versatile Types of Organic/Inorganic Nanohybrids: From Strategic Design to Biomedical Applications. Chem Rev 2018; 119:1666-1762. [DOI: 10.1021/acs.chemrev.8b00401] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Nana Zhao
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Liemei Yan
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoyi Zhao
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xinyan Chen
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Aihua Li
- College of Materials Science and Engineering, Institute for Graphene Applied Technology Innovation, Laboratory of Fiber Materials and Modern Textiles, Growing Base for State Key Laboratory, Collaborative Innovation Center for Marine Biomass Fibers Materials and Textiles of Shandong Province, Qingdao University, Qingdao 266071, China
| | - Di Zheng
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xin Zhou
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoguang Dai
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Beijing Laboratory of Biomedical Materials, Key Laboratory of Carbon Fiber and Functional Polymers (Beijing University of Chemical Technology), Ministry of Education, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
45
|
Chen W, Cheng CA, Lee BY, Clemens DL, Huang WY, Horwitz MA, Zink JI. Facile Strategy Enabling Both High Loading and High Release Amounts of the Water-Insoluble Drug Clofazimine Using Mesoporous Silica Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2018; 10:31870-31881. [PMID: 30160469 DOI: 10.1021/acsami.8b09069] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The use of nanocarriers to deliver poorly soluble drugs to the sites of diseases is an attractive and general method, and mesoporous silica nanoparticles (MSNs) are increasingly being used as carriers. However, both loading a large amount of drugs into the pores and still being able to release the drug is a challenge. In this paper, we demonstrate a general strategy based on a companion molecule that chaperones the drug into the pores and also aids it in escaping. A common related strategy is to use a miscible co-solvent dimethyl sulfoxide (DMSO), but although loading may be efficient in DMSO, this co-solvent frequently diffuses into an aqueous environment, leaving the drug behind. We demonstrate the method by using acetophenone (AP), an FDA-approved food additive as the chaperone for clofazimine (CFZ), a water-insoluble antibiotic used to treat leprosy and multidrug-resistant tuberculosis. AP enables a high amount of CFZ cargo into the MSNs and also carries CFZ cargo out from the MSNs effectively when they are in an aqueous biorelevant environment. The amount of loading and the CFZ release efficiency from MSNs were optimized; 4.5 times more CFZ was loaded in MSNs with AP than that with DMSO and 2300 times more CFZ was released than that without the assistance of the AP. In vitro treatment of macrophages infected by Mycobacterium tuberculosis with the optimized CFZ-loaded MSNs killed the bacteria in the cells in a dose-dependent manner. These studies demonstrate a highly efficient method for loading nanoparticles with water-insoluble drug molecules and the efficacy of the nanoparticles in delivering drugs into eukaryotic cells in aqueous media.
Collapse
|
46
|
Nkanga CI, Walker RB, Krause RW. pH-Dependent release of isoniazid from isonicotinic acid (4-hydroxy-benzylidene)-hydrazide loaded liposomes. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
47
|
Nkanga CI, Krause RWM. Conjugation of isoniazid to a zinc phthalocyanine via hydrazone linkage for pH-dependent liposomal controlled release. APPLIED NANOSCIENCE 2018. [DOI: 10.1007/s13204-018-0776-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
48
|
Li D, Li L, Ma Y, Zhuang Y, Li D, Shen H, Wang X, Yang F, Ma Y, Wu D. Dopamine-assisted fixation of drug-loaded polymeric multilayers to osteoarticular implants for tuberculosis therapy. Biomater Sci 2018; 5:730-740. [PMID: 28218329 DOI: 10.1039/c7bm00042a] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Currently, the major issues in the treatment of osteoarticular tuberculosis (TB) after implant placement are low drug concentration at the infected focus and drug resistance resulting from the long-term chemotherapy. The application of drug-loaded polymeric multilayers on implantable devices offers a promising solution to the problems. Herein, a poly(ethylene glycol)-based hydrogel film embedded with isoniazid (INH)-loaded alginate microparticles was fixed to Ti implants via adhesive polydopamine, subsequently capped by poly(lactic-co-glycolic acid) membranes for the sustained and localized delivery of the anti-TB drug. The antibacterial efficacy of the released INH was confirmed by a 4.5 ± 0.8 cm inhibition zone formed in the fourth week after inoculation of Mycobacterium tuberculosis. The INH-loaded Ti implants showed no toxicity to the osteoblast cell and provided a consistent drug release for nearly one week in vitro. The release profile in vivo showed a high local concentration and low systemic exposure. The local INH concentration could be kept higher than its minimum inhibitory concentration over a period of 8 weeks, which proves that it is a promising strategy to improve the severe osteoarticular TB treatment.
Collapse
Affiliation(s)
- Dan Li
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Litao Li
- Department of Orthopaedics, The 309th Hospital of the PLA, Beijing 100094, China.
| | - Yunlong Ma
- Department of Orthopaedics, The 309th Hospital of the PLA, Beijing 100094, China.
| | - Yaping Zhuang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dawei Li
- Department of Orthopaedics, The 309th Hospital of the PLA, Beijing 100094, China.
| | - Hong Shen
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Xing Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.
| | - Fei Yang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanzheng Ma
- Department of Orthopaedics, The 309th Hospital of the PLA, Beijing 100094, China.
| | - Decheng Wu
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. and University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
49
|
Tao Y, Liu S, Zhang Y, Chi Z, Xu J. A pH-responsive polymer based on dynamic imine bonds as a drug delivery material with pseudo target release behavior. Polym Chem 2018. [DOI: 10.1039/c7py02108a] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In this study, pentaerythritol tetra(3-mercaptopropionate)-allylurea-poly(ethylene glycol) (PETMP-AU-PEG), produced by the Schiff-base reaction between terminal-aldehyded PEG and PETMP-AU, was used to prepare doxorubicin (DOX)-loaded polymers for triggered release.
Collapse
Affiliation(s)
- Yangchun Tao
- PCFM Lab and GD HPPC Lab
- School of Chemistry
- Sun Yat-sen University
- Guangzhou
- China
| | - Siwei Liu
- PCFM Lab and GD HPPC Lab
- School of Chemistry
- Sun Yat-sen University
- Guangzhou
- China
| | - Yi Zhang
- PCFM Lab and GD HPPC Lab
- School of Chemistry
- Sun Yat-sen University
- Guangzhou
- China
| | - Zhenguo Chi
- PCFM Lab and GD HPPC Lab
- School of Chemistry
- Sun Yat-sen University
- Guangzhou
- China
| | - Jiarui Xu
- PCFM Lab and GD HPPC Lab
- School of Chemistry
- Sun Yat-sen University
- Guangzhou
- China
| |
Collapse
|
50
|
Mebert AM, Baglole CJ, Desimone MF, Maysinger D. Nanoengineered silica: Properties, applications and toxicity. Food Chem Toxicol 2017; 109:753-770. [DOI: 10.1016/j.fct.2017.05.054] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 05/26/2017] [Indexed: 02/06/2023]
|