1
|
Mohanty DL, Divya N, Zafar A, Warsi MH, Parida GR, Padhi P, Khalid M, Yasir M, Mujtaba MA. Development of etoricoxib-loaded mesoporous silica nanoparticles laden gel as vehicle for transdermal delivery: optimization, ex vivo permeation, histopathology, and in vivo anti-inflammatory study. Drug Dev Ind Pharm 2025; 51:506-521. [PMID: 40192336 DOI: 10.1080/03639045.2025.2490287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/22/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
OBJECTIVE Etoricoxib (ETB) is a nonsteroidal anti-inflammatory therapeutic agent. It is poorly soluble and has various gastrointestinal side effects such as bleeding and ulcers after oral administration. The present research aimed to develop an ETB-loaded mesoporous silica nanoparticle-laden gel (ETB-MSNPs) for transdermal delivery to improve therapeutic efficacy. METHODS The ETB-MSNPs were synthesized using a precipitation and solvent evaporation technique and their optimization was performed using a Box-Behnken design. The optimized ETB-MSNPs were incorporated into a carbopol-chitosan gel and evaluated for in vitro, ex vivo, and in vivo anti-inflammatory activity. RESULTS The ETB-MSNPs displayed nanosize of particles with nanosize distribution and high entrapment efficiency of ETB. The Fourier transform infrared spectroscopy (FTIR) and differential scanning calorimetry (DSC) studies showed that ETB was encapsulated in MSNPs. The optimized ETB-MSNPs were successfully integrated into the carbopol and chitosan gel, which exhibited excellent viscosity and spreadability. The optimized ETB-MSNPs gel exhibited a significantly higher and more sustained release of ETB compared to pure ETB gel. Optimized ETB-MSNPs gel exhibited a considerably higher anti-inflammatory effect with a significant reduction in IL-1β and TNF-α levels compared to pure ETB gel. The histopathological examination confirmed that optimized ETB-MSNPs gel did not exhibit any toxicity on the skin. CONCLUSION Based on the findings, the results suggest that the MSNPs gel has the potential as a carrier for enhancing the therapeutic efficacy of ETB through topical delivery, although further studies are needed to fully confirm its effectiveness.
Collapse
Affiliation(s)
- Dibya Lochan Mohanty
- Department of Pharmaceutics, School of Pharmacy, Centre for Nanomedicine, Anurag University, Hyderabad, India
| | - Noota Divya
- Department of Pharmaceutics, School of Pharmacy, Centre for Nanomedicine, Anurag University, Hyderabad, India
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Musarrat Husain Warsi
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Gnyana Ranjan Parida
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar, India
| | - Priyanka Padhi
- Gayatri Institute of Science and Technology, Gunupur, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohd Yasir
- Department of Pharmacy, College of Health Sciences, Arsi University, Asella, Ethiopia
| | - Md Ali Mujtaba
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Arar, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
| |
Collapse
|
2
|
Agrawal SS, Baliga V, Londhe VY. Liposomal Formulations: A Recent Update. Pharmaceutics 2024; 17:36. [PMID: 39861685 PMCID: PMC11769406 DOI: 10.3390/pharmaceutics17010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 01/27/2025] Open
Abstract
Liposome-based drug delivery technologies have showed potential in enhancing medication safety and efficacy. Innovative drug loading and release mechanisms highlighted in this review of next-generation liposomal formulations. Due to poor drug release kinetics and loading capacity, conventional liposomes have limited clinical use. Scientists have developed new liposomal carrier medication release control and encapsulation methods to address these limits. Drug encapsulation can be optimized by creating lipid compositions that match a drug's charge and hydrophobicity. By selecting lipids and adding co-solvents or surfactants, scientists have increased drug loading in liposomal formulations while maintaining stability. Nanotechnology has also created multifunctional liposomes with triggered release and personalized drug delivery. Surface modification methods like PEGylation and ligand conjugation can direct liposomes to disease regions, improving therapeutic efficacy and reducing off-target effects. In addition to drug loading, researchers have focused on spatiotemporal modulation of liposomal carrier medication release. Stimuli-responsive liposomes release drugs in response to bodily signals. Liposomes can be pH- or temperature-sensitive. To improve therapeutic efficacy and reduce systemic toxicity, researchers added stimuli-responsive components to liposomal membranes to precisely control drug release kinetics. Advanced drug delivery technologies like magnetic targeting and ultrasound. Pro Drug, RNA Liposomes approach may improve liposomal medication administration. Magnetic targeting helps liposomes aggregate at illness sites and improves drug delivery, whereas ultrasound-mediated drug release facilitates on-demand release of encapsulated medicines. This review also covers recent preclinical and clinical research showing the therapeutic promise of next-generation liposomal formulations for cancer, infectious diseases, neurological disorders and inflammatory disorders. The transfer of these innovative liposomal formulations from lab to clinical practice involves key difficulties such scalability, manufacturing difficulty, and regulatory limits.
Collapse
Affiliation(s)
- Surendra S. Agrawal
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (DU), Sawangi (M), Wardha 442001, Maharashtra, India;
| | - Vrinda Baliga
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India
| | - Vaishali Y. Londhe
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India
| |
Collapse
|
3
|
Aljabali AAA, Obeid MA, Gammoh O, El-Tanani M, Mishra V, Mishra Y, Kapre S, Srivatsa Palakurthi S, Hassan SS, Nawn D, Lundstrom K, Hromić-Jahjefendić A, Serrano-Aroca Á, Redwan EM, Uversky VN, Tambuwala MM. Nanomaterial-Driven Precision Immunomodulation: A New Paradigm in Therapeutic Interventions. Cancers (Basel) 2024; 16:2030. [PMID: 38893150 PMCID: PMC11171400 DOI: 10.3390/cancers16112030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Immunotherapy is a rapidly advancing field of research in the treatment of conditions such as cancer and autoimmunity. Nanomaterials can be designed for immune system manipulation, with precise targeted delivery and improved immunomodulatory efficacy. Here, we elaborate on various strategies using nanomaterials, including liposomes, polymers, and inorganic NPs, and discuss their detailed design intricacies, mechanisms, and applications, including the current regulatory issues. This type of nanomaterial design for targeting specific immune cells or tissues and controlling release kinetics could push current technological frontiers and provide new and innovative solutions for immune-related disorders and diseases without off-target effects. These materials enable targeted interactions with immune cells, thereby enhancing the effectiveness of checkpoint inhibitors, cancer vaccines, and adoptive cell therapies. Moreover, they allow for fine-tuning of immune responses while minimizing side effects. At the intersection of nanotechnology and immunology, nanomaterial-based platforms have immense potential to revolutionize patient-centered immunotherapy and reshape disease management. By prioritizing safety, customization, and compliance with regulatory standards, these systems can make significant contributions to precision medicine, thereby significantly impacting the healthcare landscape.
Collapse
Affiliation(s)
- Alaa A. A. Aljabali
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan; (A.A.A.A.); (M.A.O.)
| | - Mohammad A. Obeid
- Faculty of Pharmacy, Department of Pharmaceutics & Pharmaceutical Technology, Yarmouk University, Irbid 21163, Jordan; (A.A.A.A.); (M.A.O.)
| | - Omar Gammoh
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Yarmouk University, Irbid 21163, Jordan;
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India;
| | - Sumedha Kapre
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.K.); (S.S.P.)
| | - Sushesh Srivatsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (S.K.); (S.S.P.)
| | - Sk. Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur 721140, West Bengal, India;
| | - Debaleena Nawn
- Indian Research Institute for Integrated Medicine (IRIIM), Unsani, Howrah 711302, West Bengal, India;
| | | | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina;
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Elrashdy M. Redwan
- Department of Biological Science, Faculty of Science, King Abdulaziz University, P.O. Box 80203, Jeddah 21589, Saudi Arabia;
- Centre of Excellence in Bionanoscience Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M. Tambuwala
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| |
Collapse
|
4
|
Panthi VK, Fairfull-Smith KE, Islam N. Liposomal drug delivery strategies to eradicate bacterial biofilms: Challenges, recent advances, and future perspectives. Int J Pharm 2024; 655:124046. [PMID: 38554739 DOI: 10.1016/j.ijpharm.2024.124046] [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: 12/23/2023] [Revised: 03/08/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Typical antibiotic treatments are often ineffectual against biofilm-related infections since bacteria residing within biofilms have developed various mechanisms to resist antibiotics. To overcome these limitations, antimicrobial-loaded liposomal nanoparticles are a promising anti-biofilm strategy as they have demonstrated improved antibiotic delivery and eradication of bacteria residing in biofilms. Antibiotic-loaded liposomal nanoparticles revealed remarkably higher antibacterial and anti-biofilm activities than free drugs in experimental settings. Moreover, liposomal nanoparticles can be used efficaciously for the combinational delivery of antibiotics and other antimicrobial compounds/peptide which facilitate, for instance, significant breakdown of the biofilm matrix, increased bacterial elimination from biofilms and depletion of metabolic activity of various pathogens. Drug-loaded liposomes have mitigated recurrent infections and are considered a promising tool to address challenges associated to antibiotic resistance. Furthermore, it has been demonstrated that surface charge and polyethylene glycol modification of liposomes have a notable impact on their antibacterial biofilm activity. Future investigations should tackle the persistent hurdles associated with development of safe and effective liposomes for clinical application and investigate novel antibacterial treatments, including CRISPR-Cas gene editing, natural compounds, phages, and nano-mediated approaches. Herein, we emphasize the significance of liposomes in inhibition and eradication of various bacterial biofilms, their challenges, recent advances, and future perspectives.
Collapse
Affiliation(s)
- Vijay Kumar Panthi
- Pharmacy Discipline, School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Kathryn E Fairfull-Smith
- School of Chemistry and Physics, Faculty of Science, Queensland University of Technology, Brisbane, Queensland 4001, Australia; Centre for Materials Science, Queensland University of Technology, Brisbane, Queensland 4001, Australia
| | - Nazrul Islam
- Pharmacy Discipline, School of Clinical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, Australia; Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane, QLD, Australia; Centre for Immunology and Infection Control (CIIC), Queensland University of Technology (QUT), Brisbane, QLD, Australia.
| |
Collapse
|
5
|
Pinho JO, Ferreira M, Coelho M, Pinto SN, Aguiar SI, Gaspar MM. Liposomal Rifabutin-A Promising Antibiotic Repurposing Strategy against Methicillin-Resistant Staphylococcus aureus Infections. Pharmaceuticals (Basel) 2024; 17:470. [PMID: 38675432 PMCID: PMC11053623 DOI: 10.3390/ph17040470] [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: 02/16/2024] [Revised: 03/23/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (M RSA) infections, in particular biofilm-organized bacteria, remain a clinical challenge and a serious health problem. Rifabutin (RFB), an antibiotic of the rifamycins class, has shown in previous work excellent anti-staphylococcal activity. Here, we proposed to load RFB in liposomes aiming to promote the accumulation of RFB at infected sites and consequently enhance the therapeutic potency. Two clinical isolates of MRSA, MRSA-C1 and MRSA-C2, were used to test the developed formulations, as well as the positive control, vancomycin (VCM). RFB in free and liposomal forms displayed high antibacterial activity, with similar potency between tested formulations. In MRSA-C1, minimal inhibitory concentrations (MIC) for Free RFB and liposomal RFB were 0.009 and 0.013 μg/mL, respectively. Minimum biofilm inhibitory concentrations able to inhibit 50% biofilm growth (MBIC50) for Free RFB and liposomal RFB against MRSA-C1 were 0.012 and 0.008 μg/mL, respectively. Confocal microscopy studies demonstrated the rapid internalization of unloaded and RFB-loaded liposomes in the bacterial biofilm matrix. In murine models of systemic MRSA-C1 infection, Balb/c mice were treated with RFB formulations and VCM at 20 and 40 mg/kg of body weight, respectively. The in vivo results demonstrated a significant reduction in bacterial burden and growth index in major organs of mice treated with RFB formulations, as compared to Control and VCM (positive control) groups. Furthermore, the VCM therapeutic dose was two fold higher than the one used for RFB formulations, reinforcing the therapeutic potency of the proposed strategy. In addition, RFB formulations were the only formulations associated with 100% survival. Globally, this study emphasizes the potential of RFB nanoformulations as an effective and safe approach against MRSA infections.
Collapse
Affiliation(s)
- Jacinta O. Pinho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.O.P.); (M.F.); (M.C.)
| | - Magda Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.O.P.); (M.F.); (M.C.)
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal
| | - Mariana Coelho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.O.P.); (M.F.); (M.C.)
| | - Sandra N. Pinto
- iBB-Institute for Bioengineering and Biosciences and Associate Laboratory i4HB−Institute for Health and Bioeconomy at Department of Bioengineering, Instituto SuperiorTécnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal;
| | - Sandra I. Aguiar
- Center for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal;
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (J.O.P.); (M.F.); (M.C.)
- IBEB, Institute of Biophysics and Biomedical Engineering, Faculty of Sciences, Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| |
Collapse
|
6
|
Ghosh R, De M. Liposome-Based Antibacterial Delivery: An Emergent Approach to Combat Bacterial Infections. ACS OMEGA 2023; 8:35442-35451. [PMID: 37810644 PMCID: PMC10551917 DOI: 10.1021/acsomega.3c04893] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023]
Abstract
The continued emergence and spread of drug-resistant pathogens and the decline in the approval of new antimicrobial drugs pose a major threat to managing infectious diseases, resulting in high morbidity and mortality. Even though a significant variety of antibiotics can effectively cure many bacterial infectious diseases, microbial infections remain one of the biggest global health problems, which may be due to the traditional drug delivery system's shortcomings which lead to poor therapeutic index, low drug absorption, and numerous other drawbacks. Further, the use of traditional antibiotics to treat infectious diseases has always been accompanied by the emergence of multidrug resistance and adverse side effects. Despite developing numerous new antibiotics, nanomaterials, and various techniques to combat infectious diseases, they have persisted as major global health issues. Improving the current antibiotic delivery systems is a promising approach to solving many life-threatening infections. In this context, nanoliposomal systems have recently attracted much attention. Herein, we attempt to provide a concise summary of recent studies that have used liposomal nanoparticles as delivery systems for antibacterial medicines. The minireview also highlights the enormous potential of liposomal nanoparticles as antibiotic delivery systems. The future of these promising approaches lies in developing more efficient delivery systems by precisely targeting bacterial cells with antibiotics with minimum cytotoxicity and high bacterial combating efficacy.
Collapse
Affiliation(s)
- Rita Ghosh
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| | - Mrinmoy De
- Department of Organic Chemistry, Indian Institute of Science, Bangalore 560012, Karnataka, India
| |
Collapse
|
7
|
de Oliveira Kocerginsky P, Dos Santos Soares PH, Lyra HFS, Cadena PG, de Lima-Neto RG, Pontes-Filho NT, Lima-Filho JVM, Costa-Júnior SD, Neves RP, Cavalcanti IMF, Santos-Magalhães NS. Efficacy and non-toxicity of ciclopirox olamine-loaded liposomes against Cryptococcus neoformans clinical isolates. Braz J Microbiol 2023; 54:1513-1521. [PMID: 37540461 PMCID: PMC10484888 DOI: 10.1007/s42770-023-01071-6] [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: 04/14/2023] [Accepted: 07/03/2023] [Indexed: 08/05/2023] Open
Abstract
The aim of this study was to evaluate the efficacy and non-toxicity of ciclopirox olamine-loaded liposomes against Cryptococcus neoformans clinical isolates. Initially, 24-1 fractional experimental design was carried out to obtain an optimized formulation of liposomes containing CPO (CPO-LipoC), which were then used to prepare stealth liposomes (CPO-LipoS). Liposomal formulations were characterized by their mean size diameter, polydispersity index (PDI), and drug encapsulation efficiency (EE%). Immunosuppressed mice were exposed to CPO-LipoS at 0.5 mg/kg/day for 14 days to verify possible histopathological alterations in the liver and kidneys. Immunosuppressed mice infected with C. neoformans were treated with CPO-LipoS at 0.5 mg/kg/day for 14 days to quantify the fungal burden in spleen, liver, lungs, and brain. CPO-LipoS presented a mean size diameter, PDI, and EE% of 101.4 ± 0.7 nm, 0.307, and 96.4 ± 0.9%, respectively. CPO-LipoS was non-toxic for the liver and kidneys of immunosuppressed mice. At the survival curve, all infected animals submitted to treatment with CPO-LipoS survived until the end of the experiment. Treatment with CPO-LipoS reduced C. neoformans cells in the spleen (59.3 ± 3.4%), liver (75.0 ± 3.6%), lungs (75.7 ± 6.7%), and brain (54.2 ± 3.2%). CPO-LipoS exhibit antifungal activity against C. neoformans, and the encapsulation of CPO into stealth liposomes allows its use as a systemic drug for treating cryptococcosis.
Collapse
Affiliation(s)
- Patrícia de Oliveira Kocerginsky
- Biosciences Center, Laboratory of Medical Mycology, Federal University of Pernambuco (UFPE), Av Reitor Joaquim Amazonas, S/N, Cidade Universitária, Recife, PE, 50740-570, Brazil
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Pedro Henrique Dos Santos Soares
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Hannah Ferreira Soares Lyra
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Pabyton Gonçalves Cadena
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
- Department of Animal Morphology and Physiology, Federal Rural University of Pernambuco (UFRPE), Rua Dom Manoel de Medeiros, S/N, Dois Irmãos, Recife, PE, 52171-900, Brazil
| | - Reginaldo Gonçalves de Lima-Neto
- Biosciences Center, Laboratory of Medical Mycology, Federal University of Pernambuco (UFPE), Av Reitor Joaquim Amazonas, S/N, Cidade Universitária, Recife, PE, 50740-570, Brazil
- Health Sciences Center, Department of Tropical Medicine, Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Nicodemos Teles Pontes-Filho
- Center for Health Sciences, Department of Pathology, Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - José Vitor Moreira Lima-Filho
- Department of Biology, Federal Rural University of Pernambuco (UFRPE), Rua Dom Manoel de Medeiros, 401, Dois Irmãos, Recife, PE, 52171-900, Brazil
| | - Sérgio Dias Costa-Júnior
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil
| | - Rejane Pereira Neves
- Biosciences Center, Laboratory of Medical Mycology, Federal University of Pernambuco (UFPE), Av Reitor Joaquim Amazonas, S/N, Cidade Universitária, Recife, PE, 50740-570, Brazil
| | - Isabella Macário Ferro Cavalcanti
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil.
- Academic Center of Vitória (CAV), Laboratory of Microbiology and Immunology, Vitória de Santo Antão, Federal University of Pernambuco (UFPE), Rua Alto Do Reservatório, S/N, Vitória de Santo Antão, PE, Brazil.
| | - Nereide Stela Santos-Magalhães
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, PE, 50670-901, Brazil.
| |
Collapse
|
8
|
Tziveleka LA, Pippa N, Ioannou E, Demetzos C, Roussis V. Development of Ulvan-Containing Liposomes as Antibacterial Drug Delivery Platforms. J Funct Biomater 2022; 13:jfb13040186. [PMID: 36278655 PMCID: PMC9589965 DOI: 10.3390/jfb13040186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/03/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Liposomes, due to their safety profile and targeting ability, are among the most studied nanocarriers as antimicrobial delivery systems. However, due to lack of stability and the non-specific interaction of liposomes with cells and proteins, their use is relatively limited. Aiming to overcome these drawbacks, it was envisaged that incorporation of ulvan, a bioactive marine sulfated polysaccharide isolated from green algae, in liposomes could improve their physicochemical properties and overall stability. Thus, we initially studied the interactions of ulvan with neutral, negatively, and positively charged lipids using Differential Scanning Calorimetry and subsequently, based on the obtained results, we prepared the respective ulvan–containing neutral and charged liposomes, where ulvan interacts with both lipid chains and polar groups in the liposomal bilayer. In a further step, we entrapped in the liposomes fusidic acid, used as a model antibacterial drug, and proceeded with the evaluation of their antibacterial activity against Staphylococcus aureus. The physicochemical properties (size and ζ-potential), stability, morphology, and entrapment efficiency of the prepared liposomal formulations were determined.
Collapse
Affiliation(s)
- Leto-Aikaterini Tziveleka
- Section of Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Natassa Pippa
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Efstathia Ioannou
- Section of Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Costas Demetzos
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
- Correspondence: (C.D.); (V.R.)
| | - Vassilios Roussis
- Section of Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
- Correspondence: (C.D.); (V.R.)
| |
Collapse
|
9
|
Maxwell A, Chaudhari BB, Chaudhari P, Ananthamurthy K, Aranjani J, Moorkoth S, Ghate V, Lewis S. In vitro antibacterial activity and in vivo pharmacokinetics of intravenously administered Amikacin-loaded Liposomes for the management of bacterial septicaemia. Colloids Surf B Biointerfaces 2022; 220:112892. [DOI: 10.1016/j.colsurfb.2022.112892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/10/2022] [Accepted: 09/28/2022] [Indexed: 10/14/2022]
|
10
|
Zouliati K, Stavropoulou P, Chountoulesi M, Naziris N, Demisli S, Mitsou E, Papadimitriou V, Chatzidaki M, Xenakis A, Demetzos C. Development and evaluation of liposomal nanoparticles incorporating dimethoxycurcumin. In vitro toxicity and permeability studies. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Monteiro KLC, Silva ON, Dos Santos Nascimento IJ, Mendonça Júnior FJB, Aquino PGV, da Silva-Júnior EF, de Aquino TM. Medicinal Chemistry of Inhibitors Targeting Resistant Bacteria. Curr Top Med Chem 2022; 22:1983-2028. [PMID: 35319372 DOI: 10.2174/1568026622666220321124452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/01/2022] [Accepted: 02/13/2022] [Indexed: 12/15/2022]
Abstract
The discovery of antibiotics was a revolutionary feat that provided countless health benefits. The identification of penicillin by Alexander Fleming initiated the era of antibiotics, represented by constant discoveries that enabled effective treatments for the different classes of diseases caused by bacteria. However, the indiscriminate use of these drugs allowed the emergence of resistance mechanisms of these microorganisms against the available drugs. In addition, the constant discoveries in the 20th century generated a shortage of new molecules, worrying health agencies and professionals about the appearance of multidrug-resistant strains against available drugs. In this context, the advances of recent years in molecular biology and microbiology have allowed new perspectives in drug design and development, using the findings related to the mechanisms of bacterial resistance to generate new drugs that are not affected by such mechanisms and supply new molecules to be used to treat resistant bacterial infections. Besides, a promising strategy against bacterial resistance is the combination of drugs through adjuvants, providing new expectations in designing new antibiotics and new antimicrobial therapies. Thus, this manuscript will address the main mechanisms of bacterial resistance under the understanding of medicinal chemistry, showing the main active compounds against efflux mechanisms, and also the application of the use of drug delivery systems, and finally, the main potential natural products as adjuvants or with promising activity against resistant strains.
Collapse
Affiliation(s)
- Kadja Luana Chagas Monteiro
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Osmar Nascimento Silva
- Faculty of Pharmacy, University Center of Anápolis, Unievangélica, 75083-515, Anápolis, Goiás, Brazil
| | - Igor José Dos Santos Nascimento
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | | | | | - Edeildo Ferreira da Silva-Júnior
- Laboratory of Medicinal Chemistry, Institute of Pharmaceutical Sciences, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, 57072-970, Maceió, Alagoas, Brazil
| |
Collapse
|
12
|
Maxwell A, Mary E, Ghate V, Aranjani J, Lewis S. A Novel high throughput 96-well based Fluorimetric Method to Measure Amikacin in Pharmaceutical Formulations: Development using Response Surface Methodology. LUMINESCENCE 2022; 37:930-943. [PMID: 35322527 DOI: 10.1002/bio.4238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 11/10/2022]
Abstract
An aminoglycoside antibiotic, amikacin, is used to treat severe and recurring bacterial infections. Due to the absence of a chromophore, however, amikacin must be extensively derivatized before being quantified, both in analytical and bioanalytical samples. In this study, for the first time, we developed a simple and sensitive method for measuring amikacin sulfate by spectrofluorimetry using a 96-well plate reader, based on the design of the experiment's approach. To develop a robust and reproducible spectrofluorimetric method, the influence of essential attributes, namely pH of the buffer, heating temperature, and concentration of reagents, were evaluated by univariate analysis followed by multivariate analysis (central composite design). ICH guidelines were used to validate the optimized method. The developed technique is linear from 1.9 to 10 μg/mL with a regression coefficient of 0.9991. The detection and quantification limits were 0.649 μg/mL and 1.9 μg/mL, respectively. For the developed method, both intra- and inter-day precision (%RSD) were below 5%. Using the method, amikacin concentrations were quantified in prepared amikacin liposomes and commercial formulations of Amicin®. The developed method greatly reduces sample volume and is a rapid, high throughput microplate-based fluorescence approach for the convenient and cost-effective measurement of amikacin in pharmaceutical formulations. In comparison to previously published approaches, the suggested method allowed for quick analysis of a high number of samples in a short amount of time (96 samples in 125 seconds), resulting in an average duration of analysis of 1.3 seconds per sample.
Collapse
Affiliation(s)
- Amala Maxwell
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Elizabeth Mary
- Department of School of Pharmacy, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana
| | - Vivek Ghate
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Jesil Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
13
|
Weldick PJ, Wang A, Halbus AF, Paunov VN. Emerging nanotechnologies for targeting antimicrobial resistance. NANOSCALE 2022; 14:4018-4041. [PMID: 35234774 DOI: 10.1039/d1nr08157h] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Antimicrobial resistance is a leading cause of mortality worldwide. Without newly approved antibiotics and antifungals being brought to the market, resistance is being developed to the ones currently available to clinicians. The reason is the applied evolutionary pressure to bacterial and fungal species due to the wide overuse of common antibiotics and antifungals in clinical practice and agriculture. Biofilms harbour antimicrobial-resistant subpopulations, which make their antimicrobial treatment even more challenging. Nanoparticle-based technologies have recently been shown to successfully overcome antimicrobial resistance in both planktonic and biofilms phenotypes. This results from the combination of novel nanomaterial research and classic antimicrobial therapies which promise to deliver a whole new generation of high-performance active nanocarrier systems. This review discusses the latest developments of promising nanotechnologies with applications against resistant pathogens and evaluates their potential and feasibility for use in novel antimicrobial therapies.
Collapse
Affiliation(s)
- Paul J Weldick
- Department of Chemistry and Biochemistry, University of Hull, Hull, HU6 7RX, UK
| | - Anheng Wang
- Department of Chemistry and Biochemistry, University of Hull, Hull, HU6 7RX, UK
| | - Ahmed F Halbus
- Department of Chemistry, College of Science, University of Babylon, Hilla, Iraq
| | - Vesselin N Paunov
- Department of Chemistry, Nazarbayev University, Kabanbay Baryr Ave. 53, Nur-sultan city, 010000, Kazakhstan.
| |
Collapse
|
14
|
Yang S, Lyu X, Zhang J, Shui Y, Yang R, Xu X. The Application of Small Molecules to the Control of Typical Species Associated With Oral Infectious Diseases. Front Cell Infect Microbiol 2022; 12:816386. [PMID: 35265531 PMCID: PMC8899129 DOI: 10.3389/fcimb.2022.816386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
Oral microbial dysbiosis is the major causative factor for common oral infectious diseases including dental caries and periodontal diseases. Interventions that can lessen the microbial virulence and reconstitute microbial ecology have drawn increasing attention in the development of novel therapeutics for oral diseases. Antimicrobial small molecules are a series of natural or synthetic bioactive compounds that have shown inhibitory effect on oral microbiota associated with oral infectious diseases. Novel small molecules, which can either selectively inhibit keystone microbes that drive dysbiosis of oral microbiota or inhibit the key virulence of the microbial community without necessarily killing the microbes, are promising for the ecological management of oral diseases. Here we discussed the research progress in the development of antimicrobial small molecules and delivery systems, with a particular focus on their antimicrobial activity against typical species associated with oral infectious diseases and the underlying mechanisms.
Collapse
Affiliation(s)
- Sirui Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoying Lyu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yusen Shui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ran Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Al Hagbani T, Rizvi SMD, Hussain T, Mehmood K, Rafi Z, Moin A, Abu Lila AS, Alshammari F, Khafagy ES, Rahamathulla M, Abdallah MH. Cefotaxime Mediated Synthesis of Gold Nanoparticles: Characterization and Antibacterial Activity. Polymers (Basel) 2022; 14:polym14040771. [PMID: 35215685 PMCID: PMC8875691 DOI: 10.3390/polym14040771] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/11/2022] Open
Abstract
Cefotaxime (CTX) is a third-generation cephalosporin antibiotic with broad-spectrum activity against Gram negative and Gram positive bacteria. However, like other third-generation cephalosporin antibiotics, its efficacy is declining due to the increased prevalence of multidrug-resistant (MDR) pathogens. Recent advances in nanotechnology have been projected as a practical approach to combat MDR microorganisms. Therefore, in the current study, gold nanoparticles (AuNPs) were prepared using cefotaxime sodium, which acted as a reducing and capping agent, besides having well-established antibacterial activity. The synthesized cefotaxime-loaded gold nanoparticles (C-AuNPs) were characterized by UV-Visible spectroscopy, FTIR, TEM and DLS. In addition, the in vitro antibacterial activity of C-AuNPs was assessed against both Gram-positive and Gram-negative bacteria. UV-Visible spectroscopy verified the formation of C-AuNPs, while TEM and DLS verified their nano-size. In addition, CTX loading onto AuNPs was confirmed by FTIR. Furthermore, the colloidal stability of the synthesized C-AuNPs was ascribed to the higher net negative surface charge of C-AuNPs. Most importantly, the synthesized C-AuNPs showed superior antibacterial activity and lower minimum inhibitory concentration (MIC) values against Gram-negative (Escherichia coli, Klebsiella oxytoca, Pseudomonas aeruginosa) and gram-positive (Staphylococcus aureus) bacteria, compared with pure CTX. Collectively, CTX was successfully adopted, as reducing and capping agent, to synthesize stable, nano-sized spherical C-AuNPs. Furthermore, loading CTX onto AuNPs could efficiently restore and/or boost the antibacterial activity of CTX against resistant Gram-negative and Gram-positive bacteria.
Collapse
Affiliation(s)
- Turki Al Hagbani
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia; (T.A.H.); (S.M.D.R.); (A.S.A.L.); (F.A.); (M.H.A.)
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia; (T.A.H.); (S.M.D.R.); (A.S.A.L.); (F.A.); (M.H.A.)
| | - Talib Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia;
| | - Khalid Mehmood
- Department of Pharmacy, Abbottabad University of Science and Technology, Havelian 22500, Pakistan;
| | - Zeeshan Rafi
- Nanomedicine and Nanotechnology Lab-6 (IIRC), Department of Biosciences, Integral University Lucknow, Lucknow 226026, India;
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia; (T.A.H.); (S.M.D.R.); (A.S.A.L.); (F.A.); (M.H.A.)
- Correspondence:
| | - Amr Selim Abu Lila
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia; (T.A.H.); (S.M.D.R.); (A.S.A.L.); (F.A.); (M.H.A.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Farhan Alshammari
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia; (T.A.H.); (S.M.D.R.); (A.S.A.L.); (F.A.); (M.H.A.)
| | - El-Sayed Khafagy
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj 11942, Saudi Arabia;
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University (KKU), Abha 61421, Saudi Arabia;
| | - Marwa H. Abdallah
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia; (T.A.H.); (S.M.D.R.); (A.S.A.L.); (F.A.); (M.H.A.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
16
|
Yu CX, Jiang W, Wang KZ, Liang AP, Song JG, Zhou YL, Sun XQ, Liu LL. Luminescent Two-Dimensional Metal-Organic Framework Nanosheets with Large π-Conjugated System: Design, Synthesis, and Detection of Anti-Inflammatory Drugs and Pesticides. Inorg Chem 2022; 61:982-991. [PMID: 34968039 DOI: 10.1021/acs.inorgchem.1c03040] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two-dimensional (2D) metal-organic framework (MOF) nanosheets, with largely exposed surface area and highly accessible active sites, have emerged as a novel kind of sensing material. Here, a luminescent 2D MOF nanosheet was designed and synthesized by a facile top-down strategy based on a three-dimensional (3D) layered MOF {[Zn(H2L)(H2O)2]·H2O}n (Zn-MOF; H4L = 3,5-bis(3',5'-dicarboxyphenyl)-1H-1,2,4-triazole). With a large π-conjugated system and rigid planar structure, ligand H4L was elaborately selected to construct the bulk Zn-MOF, which can be readily exfoliated into 2D nanosheets, owing to the weak interlayer interactions and easy-to-release H2O molecules in the interspaces of 2D layers. Given the great threat posed to the ecological environment by anti-inflammatory drugs and pesticides, the developed luminescent Zn-MOF nanosheets were utilized to determine these organic pollutants, achieving highly selective and sensitive detection of diclofenac sodium (DCF) and tetramethylthiuram disulfide (TMTD). Compared to the detection limits of 3D Zn-MOF (7.72 ppm for DCF, 6.01 ppm for TMTD), the obviously lower detection limits for 2D Zn-MOF nanosheets toward DCF (0.20 ppm) and TMTD (0.18 ppm) further revealed that the largely exposed surface area with rigid planar structure and ultralarge π-conjugated system greatly accelerated electron transfer, which brought about a vast improvement in response sensitivity. The remarkable quenching performance for DCF and TMTD stems from a combined effect of photoinduced electron transfer and competitive energy absorption. The possible sensing mechanism was systematically investigated by the studies of powder X-ray diffraction, UV-vis, luminescence lifetime, and density functional theory calculations.
Collapse
Affiliation(s)
- Cai-Xia Yu
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| | - Wen Jiang
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| | - Ke-Zhong Wang
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| | - Ai-Ping Liang
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| | - Jian-Guo Song
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| | - Yan-Li Zhou
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| | - Xue-Qin Sun
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| | - Lei-Lei Liu
- School of Environmental and Material Engineering, Yantai University, Yantai 264005, P.R. China
| |
Collapse
|
17
|
Meers PR. Membrane Organization Strategies in Vesicular Antibiotic Delivery. J Membr Biol 2022; 255:523-535. [PMID: 35018488 DOI: 10.1007/s00232-021-00210-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 01/08/2023]
Abstract
Small molecule antibiotics are often derived from microorganisms that thrive in competitive environments. Their importance as therapeutics for infectious disease in humans has been established over many years. It has now become clear that antibiotic-producing organisms use packaging and delivery in the form of vesicles in many cases. A similar strategy has evolved in recent decades in the pharmaceutical industry for formulation of antibiotic therapies. The top-down approach that has evolved over millions of years in various micro-organisms has generated complex, efficient delivery systems that we are just now beginning to understand. The bottom-up formulation approach involves simple, safe compositions, which are being continually enhanced by trying to add features of which the natural systems inform us. A comparison is made here of these paradigms. Despite the differences, there are a number of common features in the basic physical and biological requirements that must be satisfied. In this review, illustration and comparison of some of these requirements is given, demonstrating the ongoing challenges in this area of research.
Collapse
Affiliation(s)
- Paul R Meers
- Department of Plant Biology, School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
18
|
Systematic review on activity of liposomal encapsulated antioxidant, antibiotics, and antiviral agents. J Liposome Res 2022; 32:340-353. [PMID: 35000548 DOI: 10.1080/08982104.2021.2024568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Reactive oxygen species (ROS) like superoxide anion, hydrogen peroxide, and hydroxyl radical, can be formed as normal products of aerobic metabolism. Overproduction or insufficient removal of ROS results in significant damage to cell structure and functions. Antioxidants applied directly and at relatively high concentrations to cellular systems are effective in protection against the damaging actions of ROS. Microorganisms including Gram-positive and negative bacteria, fungi, protozoa, algae, etc., can be disease causing microorganism. Antimicrobial agents have the capability to inhibitor destroy the microorganisms. The problems arising from the use of antioxidant and antimicrobial agents include poor solubility, instability during storage, low bioavailability, and difficulty to reach target organs with sufficient doses. Liposomal antimicrobial agent and liposomal antioxidants enhance the solubility, bioavailability, and stability of antimicrobial agent and antioxidants.
Collapse
|
19
|
Drost M, Diamanti E, Fuhrmann K, Goes A, Shams A, Haupenthal J, Koch M, Hirsch AKH, Fuhrmann G. Bacteriomimetic Liposomes Improve Antibiotic Activity of a Novel Energy-Coupling Factor Transporter Inhibitor. Pharmaceutics 2021; 14:4. [PMID: 35056900 PMCID: PMC8779172 DOI: 10.3390/pharmaceutics14010004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 11/21/2022] Open
Abstract
Liposomes have been studied for decades as nanoparticulate drug delivery systems for cytostatics, and more recently, for antibiotics. Such nanoantibiotics show improved antibacterial efficacy compared to the free drug and can be effective despite bacterial recalcitrance. In this work, we present a loading method of bacteriomimetic liposomes for a novel, hydrophobic compound (HIPS5031) inhibiting energy-coupling factor transporters (ECF transporters), an underexplored antimicrobial target. The liposomes were composed of DOPG (18:1 (Δ9-cis) phosphatidylglycerol) and CL (cardiolipin), resembling the cell membrane of Gram-positive Staphylococcus aureus and Streptococcus pneumoniae, and enriched with cholesterol (Chol). The size and polydispersity of the DOPG/CL/± Chol liposomes remained stable over 8 weeks when stored at 4 °C. Loading of the ECF transporter inhibitor was achieved by thin film hydration and led to a high encapsulation efficiency of 33.19% ± 9.5% into the DOPG/CL/Chol liposomes compared to the phosphatidylcholine liposomes (DMPC/DPPC). Bacterial growth inhibition assays on the model organism Bacillus subtilis revealed liposomal HIPS5031 as superior to the free drug, showing a 3.5-fold reduction in CFU/mL at a concentration of 9.64 µM. Liposomal HIPS5031 was also shown to reduce B. subtilis biofilm. Our findings present an explorative basis for bacteriomimetic liposomes as a strategy against drug-resistant pathogens by surpassing the drug-formulation barriers of innovative, yet unfavorably hydrophobic, antibiotics.
Collapse
Affiliation(s)
- Menka Drost
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Staudtstr. 5, 91058 Erlangen, Germany
| | - Eleonora Diamanti
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
- Helmholtz International Lab for Anti-Infectives, Campus E8.1, 66123 Saarbrücken, Germany
| | - Kathrin Fuhrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
| | - Adriely Goes
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
| | - Atanaz Shams
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
| | - Jörg Haupenthal
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
| | - Marcus Koch
- INM-Leibniz-Institut für Neue Materialien, Campus D2.2, 66123 Saarbrücken, Germany;
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
- Helmholtz International Lab for Anti-Infectives, Campus E8.1, 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, Campus C1.7, 66123 Saarbrücken, Germany
| | - Gregor Fuhrmann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Campus E8.1, 66123 Saarbrücken, Germany; (M.D.); (E.D.); (K.F.); (A.G.); (A.S.); (J.H.); (A.K.H.H.)
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Staudtstr. 5, 91058 Erlangen, Germany
| |
Collapse
|
20
|
Antibacterial toxicity of mesoporous silica nanoparticles with functional decoration of specific organic moieties. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2021.127612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Nwabuife JC, Pant AM, Govender T. Liposomal delivery systems and their applications against Staphylococcus aureus and Methicillin-resistant Staphylococcus aureus. Adv Drug Deliv Rev 2021; 178:113861. [PMID: 34242712 DOI: 10.1016/j.addr.2021.113861] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/16/2022]
Abstract
Liposomal delivery systems have been widely explored for targeting superbugs such as S. aureus and MRSA, overcoming antimicrobial resistance associated with conventional dosage forms. They have the significant advantage of delivering hydrophilic and lipophilic antimicrobial agents, either singularly as monotherapy or in combination as combination therapy, due to their bilayers with action-site-specificity, resulting in improved targeting compared to conventional dosage forms. Herein, we present an extensive and critical review of the different liposomal delivery systems employed in the past two decades for the delivery of both antibiotics of different classes and non-antibiotic antibacterial agents, as monotherapy and combination therapy to eradicate infections caused by S. aureus and MRSA. The review also identifies future research and strategies potentiating the applications of liposomal delivery systems against S. aureus and MRSA. This review confirms the potential application of liposomal delivery systems for effective delivery and specific targeting of S. aureus and MRSA infections.
Collapse
|
22
|
Maxwell A, Ghate V, Aranjani J, Lewis S. Breaking the barriers for the delivery of amikacin: Challenges, strategies, and opportunities. Life Sci 2021; 284:119883. [PMID: 34390724 DOI: 10.1016/j.lfs.2021.119883] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/30/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022]
Abstract
Hypodermic delivery of amikacin is a widely adopted treatment modality for severe infections, including bacterial septicemia, meningitis, intra-abdominal infections, burns, postoperative complications, and urinary tract infections in both paediatric and adult populations. In most instances, the course of treatment requires repeated bolus doses of amikacin, prolonged hospitalization, and the presence of a skilled healthcare worker for administration and continuous therapeutic monitoring to manage the severe adverse effects. Amikacin is hydrophilic and exhibits a short half-life, which further challenges the delivery of sufficient systemic concentrations when administered by the oral or transdermal route. In this purview, the exploitation of novel controlled and sustained release drug delivery platforms is warranted. Furthermore, it has been shown that novel delivery systems are capable of increasing the antibacterial activity of amikacin at lower doses when compared to the conventional formulations and also aid in overcoming the development of drug-resistance, which currently is a significant threat to the healthcare system worldwide. The current review presents a comprehensive overview of the developmental history of amikacin, the mechanism of action in virulent strains as well as the occurrence of resistance, and various emerging drug delivery solutions developed both by the academia and the industry. The examples outlined within the review provides significant pieces of evidence on novel amikacin formulations in the field of antimicrobial research paving the path for future therapeutic interventions that will result in improved clinical outcome.
Collapse
Affiliation(s)
- Amala Maxwell
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Vivek Ghate
- Mechatronics Lab, Department of Electronic System Engineering, Indian Institute of Science, Bengaluru 560012, Karnataka, India
| | - Jesil Aranjani
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India.
| |
Collapse
|
23
|
Bindini E, Ramirez MDLA, Rios X, Cossío U, Simó C, Gomez-Vallejo V, Soler-Illia G, Llop J, Moya SE. In Vivo Tracking of the Degradation of Mesoporous Silica through 89 Zr Radio-Labeled Core-Shell Nanoparticles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2101519. [PMID: 34145769 DOI: 10.1002/smll.202101519] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/02/2021] [Indexed: 06/12/2023]
Abstract
While mesoporous silica nanoparticles (MSNs) are extensively studied as high-potential drug delivery platforms, the successful clinical translation of these nanocarriers strongly depends on their biodistribution, biodegradation, and elimination patterns in vivo. Here, a novel method is reported to follow the in vivo degradation of MSNs by tracking a radioactive label embedded in the silica structure. Core-shell silica nanoparticles (NPs) with a dense core and a mesoporous shell are labeled with low quantities of the positron emitter 89 Zr, either in the dense core or in the mesoporous shell. In vivo positron emission tomography imaging and ex vivo organ measurements reveal a remarkable difference in the 89 Zr biodistribution between the shell-labeled and the core-labeled NPs. Release of the radiotracer from shell-labeled NPs is used as a probe of the extent of silica dissolution, and a prompt release of the radioisotope is observed, with partial excretion already in the first 2 h post injection, and a slower accumulation in bones over time. On the other hand, when 89 Zr is embedded in the nanoparticle core, the biodistribution remains largely unchanged during the first 6 h. These findings indicate that MSNs have fast, hour-scale, degradation kinetics in vivo.
Collapse
Affiliation(s)
- Elisa Bindini
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
| | - Maria de Los Angeles Ramirez
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
- Instituto de Nanosistemas, UNSAM, CONICET, Avenida 25 de Mayo 1021, San Martín, Buenos Aires, 1650, Argentina
| | - Xabier Rios
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
| | - Unai Cossío
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
| | - Cristina Simó
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
| | - Vanessa Gomez-Vallejo
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
| | - Galo Soler-Illia
- Instituto de Nanosistemas, UNSAM, CONICET, Avenida 25 de Mayo 1021, San Martín, Buenos Aires, 1650, Argentina
| | - Jordi Llop
- Radiochemistry and Nuclear Imaging Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
- Centro de Investigación Biomédica en Red - Enfermedades Respiratorias (CIBERES), Av. Monforte de Lemos, 3-5, Madrid, 28029, Spain
| | - Sergio E Moya
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, San Sebastián, Guipúzcoa, 20014, Spain
| |
Collapse
|
24
|
Singh SK, Kumar U, Guleria A, Kumar D. A brief overview about the use of different bioactive liposome-based drug delivery systems in Peritoneal Dialysis and some other diseases. NANO EXPRESS 2021. [DOI: 10.1088/2632-959x/abfdd1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Abstract
Peritoneal dialysis (PD) is a promising way of treatment used for patients suffering from End-Stage Renal Failure (ESRF). Liposomes are nanocarriers comprised of lipid bilayers encapsulating an aqueous core. Liposomes are extensively used as drug delivery systems and several liposomal nanomedicines have been approved for clinical applications. Nanomedicine constitutes a new direction in peritonitis prevention using peritoneal dialysis (PD). In case of PD; there is a more risk of bacterial infection in the peritoneal cavity along with subcutaneous tunnel and catheter existing site. These infections are the most common complications associated with prolonged peritoneal dialysis (PD) therapy. To prevent such complications, patients used to treat with suitable antibiotic. Nanocarriers consist of assembly of nano-sized vehicles planned to deliver encapsulated/loaded bioactive(s) to the specific target (tissues or organs) and have provided prominent improved therapeutic efficacy for PD patients. The advantage of bioactive loaded nanocarrier has the efficient capacity to deliver at target specific site in PD. This review focuses mainly on the current use of different liposomal encapsulated bioactive compounds in drug delivery systems in the case of PD and other human diseases and briefly highlights the importance and use of different liposomal encapsulated antimicrobial agents to improve the PD technique.
Collapse
|
25
|
Ferreira M, Ogren M, Dias JNR, Silva M, Gil S, Tavares L, Aires-da-Silva F, Gaspar MM, Aguiar SI. Liposomes as Antibiotic Delivery Systems: A Promising Nanotechnological Strategy against Antimicrobial Resistance. Molecules 2021; 26:2047. [PMID: 33918529 PMCID: PMC8038399 DOI: 10.3390/molecules26072047] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/17/2022] Open
Abstract
Antimicrobial drugs are key tools to prevent and treat bacterial infections. Despite the early success of antibiotics, the current treatment of bacterial infections faces serious challenges due to the emergence and spread of resistant bacteria. Moreover, the decline of research and private investment in new antibiotics further aggravates this antibiotic crisis era. Overcoming the complexity of antimicrobial resistance must go beyond the search of new classes of antibiotics and include the development of alternative solutions. The evolution of nanomedicine has allowed the design of new drug delivery systems with improved therapeutic index for the incorporated compounds. One of the most promising strategies is their association to lipid-based delivery (nano)systems. A drug's encapsulation in liposomes has been demonstrated to increase its accumulation at the infection site, minimizing drug toxicity and protecting the antibiotic from peripheral degradation. In addition, liposomes may be designed to fuse with bacterial cells, holding the potential to overcome antimicrobial resistance and biofilm formation and constituting a promising solution for the treatment of potential fatal multidrug-resistant bacterial infections, such as methicillin resistant Staphylococcus aureus. In this review, we aim to address the applicability of antibiotic encapsulated liposomes as an effective therapeutic strategy for bacterial infections.
Collapse
Affiliation(s)
- Magda Ferreira
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria Ogren
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Joana N. R. Dias
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Marta Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Solange Gil
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Luís Tavares
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Frederico Aires-da-Silva
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| | - Maria Manuela Gaspar
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Sandra Isabel Aguiar
- Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; (M.F.); (M.O.); (J.N.R.D.); (M.S.); (S.G.); (L.T.); (F.A.-d.-S.)
| |
Collapse
|
26
|
Ferreira M, Pinto SN, Aires-da-Silva F, Bettencourt A, Aguiar SI, Gaspar MM. Liposomes as a Nanoplatform to Improve the Delivery of Antibiotics into Staphylococcus aureus Biofilms. Pharmaceutics 2021; 13:pharmaceutics13030321. [PMID: 33801281 PMCID: PMC7999762 DOI: 10.3390/pharmaceutics13030321] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus biofilm-associated infections are a major public health concern. Current therapies are hampered by reduced penetration of antibiotics through biofilm and low accumulation levels at infected sites, requiring prolonged usage. To overcome these, repurposing antibiotics in combination with nanotechnological platforms is one of the most appealing fast-track and cost-effective approaches. In the present work, we assessed the potential therapeutic benefit of three antibiotics, vancomycin, levofloxacin and rifabutin (RFB), through their incorporation in liposomes. Free RFB displayed the utmost antibacterial effect with MIC and MBIC50 below 0.006 µg/mL towards a methicillin susceptible S. aureus (MSSA). RFB was selected for further in vitro studies and the influence of different lipid compositions on bacterial biofilm interactions was evaluated. Although positively charged RFB liposomes displayed the highest interaction with MSSA biofilms, RFB incorporated in negatively charged liposomes displayed lower MBIC50 values in comparison to the antibiotic in the free form. Preliminary safety assessment on all RFB formulations towards osteoblast and fibroblast cell lines demonstrated that a reduction on cell viability was only observed for the positively charged liposomes. Overall, negatively charged RFB liposomes are a promising approach against biofilm S. aureus infections and further in vivo studies should be performed.
Collapse
Affiliation(s)
- Magda Ferreira
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.F.); (F.A.-d.-S.)
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Sandra N. Pinto
- Department of Bioengineering, iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal;
| | - Frederico Aires-da-Silva
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.F.); (F.A.-d.-S.)
| | - Ana Bettencourt
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Sandra I. Aguiar
- Centre for Interdisciplinary Research in Animal Health (CIISA), Faculty of Veterinary Medicine, Universidade de Lisboa, 1300-477 Lisboa, Portugal; (M.F.); (F.A.-d.-S.)
- Correspondence: (S.I.A.); (M.M.G.)
| | - Maria Manuela Gaspar
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Correspondence: (S.I.A.); (M.M.G.)
| |
Collapse
|
27
|
Karpuz M, Atlihan-Gundogdu E, Demir ES, Senyigit Z. Radiolabeled Tedizolid Phosphate Liposomes for Topical Application: Design, Characterization, and Evaluation of Cellular Binding Capacity. AAPS PharmSciTech 2021; 22:62. [PMID: 33528714 DOI: 10.1208/s12249-020-01917-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/22/2020] [Indexed: 11/30/2022] Open
Abstract
Nowadays, the incidence of acute bacterial skin and skin structure infection (ABSSSI) is increasing. The increased bioavailability and reduced drug resistance of antibiotics are crucial to obtain a more effective treatment response in these infections. These favorable properties could be achieved by different drug delivery systems such as liposomes. In this study, nanosized, radiolabeled tedizolid phosphate liposomal formulations were prepared and evaluated with their in vitro cellular binding capacity and biocompatible profile for topical treatment of ABSSSI. Liposomes were characterized by evaluation of their visual inspection, particle size (about 190-270 nm), zeta potential value (around 0), and encapsulation efficiency (nearly 10%). The release rate of tedizolid phosphate from liposomes was also studied using dialysis membranes and evaluated kinetically. The stability of formulations was observed at three different temperatures and humidity conditions for 28 days. Afterward, liposomes were labeled with 99mTc, and the optimal amount of reducing agent (stannous chloride) was determined as 500 μg in this direct labeling procedure. All liposome formulations were successfully radiolabeled with high efficiency and exhibited high radiochemical purity (> 80%) during 6 h in different media. Furthermore, the cellular bindings of liposomal formulations were evaluated in human skin fibroblast cells by measuring the radioactivity. Higher radioactivity values were obtained in CCD-1070Sk cells incubated by liposome formulations compared to sodium pertechnetate. This finding suggested that liposomal formulation increased the cellular binding of radioactivity. By the result of our study, nanosized, tedizolid phosphate encapsulated liposome formulation was found to be a favorable carrier system in the treatment of ABSSSI.
Collapse
|
28
|
Karpuz M, Silindir-Gunay M, Ozer AY, Ozturk SC, Yanik H, Tuncel M, Aydin C, Esendagli G. Diagnostic and therapeutic evaluation of folate-targeted paclitaxel and vinorelbine encapsulating theranostic liposomes for non-small cell lung cancer. Eur J Pharm Sci 2020; 156:105576. [PMID: 32987115 DOI: 10.1016/j.ejps.2020.105576] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022]
Abstract
NSCLC is the most common type of lung cancer. However, non-specific contrast agents, radiopharmaceuticals, and treatment methods are insufficient in early diagnosis and eradication of all tumor tissue. Therefore, the formulation of a novel, targeted, specific theranostic agents possess critical importance. In our previous study, paclitaxel and vinorelbine encapsulating, Tc-99m radiolabeled, folate targeted, nanosized liposomes were formulated and found promising due to characterization properties, high cellular uptake, and cytotoxicity. In this study, in vivo therapeutic and diagnostic efficacy of liposomal formulations were tested by biodistribution study, evaluation of tumor growth inhibition, and histopathologic examination after in vitro assays on LLC1 cells. Both actively and passively targeted liposomal formulations exhibited high cellular uptake, and co-drug encapsulating liposomes showed a greater cytotoxicity profiles than free drug combination in LLC1 cells. By the results of biodistribution studies performed in NSCLC tumor-bearing C57BL/6 mice, the uptake of radiolabeled, actively folate targeted, co-drug encapsulating liposomal formulation was found to be higher in tumor tissue when compared to non-actively targeted one. Also, more effective treatment was achieved by using folate-targeted, co-drug encapsulating liposomal formulation when compared to free drugs combination according to changes in tumor size of mice. Furthermore, liposomal formulations showed lower toxicity compared to free drug combinations in the toxicity study considering body weight. Moreover, according to the histopathological study, folate targeted, co-drug encapsulating liposomes not only inhibited the tumor growth effectively but also restricted the lung metastasis entirely.
Collapse
Affiliation(s)
- Merve Karpuz
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey; Department of Radiopharmacy, Faculty of Pharmacy, Izmir KatipCelebi University, Izmir, Turkey
| | - Mine Silindir-Gunay
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - A Yekta Ozer
- Department of Radiopharmacy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| | - Suleyman Can Ozturk
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Hamdullah Yanik
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Murat Tuncel
- Department of Nuclear Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Cisel Aydin
- Department of Pathology, Faculty of Medicine, Koc University, Istanbul, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
29
|
He K, Liu J, Gao Y, Hao Y, Yang X, Huang G. Preparation and Evaluation of Stearylamine-Bearing Pemetrexed Disodium-Loaded Cationic Liposomes In Vitro and In Vivo. AAPS PharmSciTech 2020; 21:193. [PMID: 32661922 DOI: 10.1208/s12249-019-1586-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/20/2019] [Indexed: 01/10/2023] Open
Abstract
Pemetrexed disodium (PMX) stands out in the treatment of non-small cell lung cancer (NSCLC), but with short half-life and toxic side effects. This study was to design cationic liposomes for targeting delivery PMX to the lungs. The PMX cationic liposome was prepared by thin-film hydration using stearylamine (SA) as the positive component of charge-regulating charge. Then, the PMX cationic liposome (SA-PMX-Lips) was characterized by particle size, morphology, entrapment efficiency (EE), and drug loading (DL). Finally, the drug release behavior in vitro, the pharmacokinetic study, and tissue distribution of SA-PMX-Lips were evaluated separately, with PMX solution (PMX-Sol) and PMX liposome (PMX-Lips) as the control. According to results, SA-PMX-Lips were spherical and the particle size was 219.7 ± 4.97 nm with a narrow polydispersity index (PDI) (0.231 ± 0.024) and a positive zeta potential 22.2 ± 0.52 mV. Its EE was 92.39 ± 1.94% and DL was 9.15 ± 0.07%. The results of in vitro and in vivo experiments showed that SA-PMX-Lips released slowly, prolonged retention time and increased the value of AUC. More notably, SA-PMX-Lips could improve the accumulation of drugs in the lungs and the relative uptake rate (Re) was 2.35 in the lungs, which indicated its lung targeting. In summary, SA-PMX-Lips showed the potential for the effective delivery of PMX and the treatment of NSCLC.
Collapse
|
30
|
Karpuz M, Silindir-Gunay M, Kursunel MA, Esendagli G, Dogan A, Ozer AY. Design and in vitro evaluation of folate-targeted, co-drug encapsulated theranostic liposomes for non-small cell lung cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101707] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
31
|
Abstract
When antibiotics are administered, orally or intravenously, they pass through different organs and layers of tissue on their way to the site of infection; this can cause dilution and/or intoxication. To overcome these problems, drug delivery vehicles have been used to encapsulate and deliver antibiotics, improving their therapeutic index while minimizing their adverse effects. Liposomes are self-assembled lipid vesicles made from at least one bilayer of phospholipids with an inner aqueous compartment. Liposomes are attractive vehicles to deliver antibiotics because they can encapsulate both hydrophobic and hydrophilic antibiotics, they have low toxicity, and they can change the biodistribution of the drug. Furthermore, liposomes have been approved by regulatory agencies. However, most developmental and mechanistic research in the field has been focused on encapsulation and delivery of anticancer drugs, a class of molecules that differ significantly in chemistry from antibiotics. In this critical Review, we discuss the state of knowledge regarding the design of liposomes for encapsulation and delivery of antibiotics and offer insight into the challenges and promises of using liposomes for antibiotic delivery.
Collapse
Affiliation(s)
- Azucena Gonzalez Gomez
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L9S 8L7, Canada
| | - Zeinab Hosseinidoust
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario L9S 8L7, Canada
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L98 4LS, Canada
| |
Collapse
|
32
|
Savadi P, Taghavi-Fard T, Milani M, Hashemzadeh N, Panahi V, McMillan NAJ, Hallaj-Nezhadi S. Piperacillin Encapsulation in Nanoliposomes Using Modified Freeze-Drying of a Monophase Solution Method: Preparation, Characterization and In Vitro Antibacterial Activity. Curr Microbiol 2020; 77:2356-2364. [PMID: 32377819 DOI: 10.1007/s00284-020-02008-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/24/2020] [Indexed: 01/16/2023]
Abstract
Piperacillin (Pip) is a broad spectrum β-lactam against most Gram-positive and Gram-negative aerobic and anaerobic bacteria. However, bacterial resistance restricts its benefits for the treatment of infectious diseases. Recently, nanoliposomal systems have been investigated as encouraging strategies to address this issue owing to their immense potential. We aimed to encapsulate Pip in liposomal nanoparticles and study their antibacterial activities in vitro against Pseudomonas aeruginosa (P. aeruginosa). Different liposomes were prepared based on the freeze-drying of a monophase solution method. Then, they were characterized in terms of size, zeta potential, polydispersity-index, and morphology. For further analysis, spectra of ATR-FTIR and XRD were taken for liposomal Pip. Encapsulation efficiency (EE) was determined via agar diffusion assay. Also, minimum inhibitory concentrations (MICs) were investigated by the standard broth macro-dilution method. The liposomes were from 100.9 to 444.13 nm with z-potential of - 30.70 to - 10.57 mV. EE of the selected formulation was 53.1%. TEM results showed that the liposomes were nanosized and almost spherical. ATR-FTIR results confirmed the full encapsulation of Pip in nanoliposomes. The X-ray pattern indicated that the liposomal Pip was amorphous. The MIC (10.6 µg/ml) of the nanoliposomal Pip against P. aeruginosa was one-half of the MIC (21.25 µg/ml) of free Pip for the same organisms. Considering four aspects (nanosized liposomes, no need for sterilization, suitable EE and enhanced antibacterial effects), this preparation method seems promising and may be used to overcome the bacterial resistance relative to Pip.
Collapse
Affiliation(s)
- Pouria Savadi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Telli Taghavi-Fard
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Milani
- Infectious and Tropical Diseases Research Center & Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahid Panahi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nigel A J McMillan
- School of Medical Sciences and Menzies Health Institute Queensland, Griffith University, Southport, Australia
| | - Somayeh Hallaj-Nezhadi
- Faculty of Pharmacy & Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Ma Y, Wang C, Li Y, Li J, Wan Q, Chen J, Tay FR, Niu L. Considerations and Caveats in Combating ESKAPE Pathogens against Nosocomial Infections. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1901872. [PMID: 31921562 PMCID: PMC6947519 DOI: 10.1002/advs.201901872] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Indexed: 05/19/2023]
Abstract
ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) are among the most common opportunistic pathogens in nosocomial infections. ESKAPE pathogens distinguish themselves from normal ones by developing a high level of antibiotic resistance that involves multiple mechanisms. Contemporary therapeutic strategies which are potential options in combating ESKAPE bacteria need further investigation. Herein, a broad overview of the antimicrobial research on ESKAPE pathogens over the past five years is provided with prospective clinical applications.
Collapse
Affiliation(s)
- Yu‐Xuan Ma
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Chen‐Yu Wang
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Yuan‐Yuan Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Jing Li
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Ji‐Hua Chen
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
| | - Franklin R. Tay
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| | - Li‐Na Niu
- State Key Laboratory of Military StomatologyNational Clinical Research Center for Oral DiseasesShaanxi Key Laboratory of StomatologyDepartment of ProsthodonticsSchool of StomatologyThe Fourth Military Medical University145 Changle West RoadXi'anShaanxi710032P. R. China
- The Graduate SchoolAugusta University1430, John Wesley Gilbert DriveAugustaGA30912‐1129USA
| |
Collapse
|
34
|
Jiang L, Lee HW, Loo SCJ. Therapeutic lipid-coated hybrid nanoparticles against bacterial infections. RSC Adv 2020; 10:8497-8517. [PMID: 35497832 PMCID: PMC9050015 DOI: 10.1039/c9ra10921h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 02/14/2020] [Indexed: 12/26/2022] Open
Abstract
One of the most important health concerns in society is the development of pathogen-causing nosocomial infections. Since the first discovery of antibiotics, bacterial infections have been highly treatable. However, with evolution and the nondiscretionary usage of antibiotics, pathogens have also found new ways to survive the onslaught of antibiotics by surviving intracellularly or through the formation of obstinate biofilms, and through these, the outcomes of regular antibiotic treatments may now be unsatisfactory. Lipid-coated hybrid nanoparticles (LCHNPs) are the next-generation core–shell structured nanodelivery system, where an inorganic or organic core, loaded with antimicrobials, is enveloped by lipid layers. This core–shell structure, with multifarious decorations, not only improves the loading capabilities of therapeutics but also has the potential to improve therapeutic delivery, especially for targeting biofilm-based and intracellular bacterial infections. Although there has been significant interest in the development of LCHNPs, they have yet to be widely exploited for bacterial infections. In this review, we will provide an overview on the latest development of LCHNPs and the various approaches in synthesizing this nano-delivery system. In addition, a discussion on future perspectives of LCHNPs, in combination with other novel anti-bacterial technologies, will be provided towards the end of this review. Lipid-coated hybrid nanoparticles are next-generation core–shell structured nanodelivery systems, which improve the loading capabilities of therapeutics and can improve therapeutic delivery, especially for targeting biofilm-based and intracellular bacterial infections.![]()
Collapse
Affiliation(s)
- Lai Jiang
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore
| | - Hiang Wee Lee
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore
| | - Say Chye Joachim Loo
- School of Materials Science & Engineering
- Nanyang Technological University
- Singapore
- Singapore Centre for Environmental Life Sciences Engineering
- Nanyang Technological University
| |
Collapse
|
35
|
Man F, Gawne PJ, T M de Rosales R. Nuclear imaging of liposomal drug delivery systems: A critical review of radiolabelling methods and applications in nanomedicine. Adv Drug Deliv Rev 2019; 143:134-160. [PMID: 31170428 PMCID: PMC6866902 DOI: 10.1016/j.addr.2019.05.012] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 12/14/2022]
Abstract
The integration of nuclear imaging with nanomedicine is a powerful tool for efficient development and clinical translation of liposomal drug delivery systems. Furthermore, it may allow highly efficient imaging-guided personalised treatments. In this article, we critically review methods available for radiolabelling liposomes. We discuss the influence that the radiolabelling methods can have on their biodistribution and highlight the often-overlooked possibility of misinterpretation of results due to decomposition in vivo. We stress the need for knowing the biodistribution/pharmacokinetics of both the radiolabelled liposomal components and free radionuclides in order to confidently evaluate the images, as they often share excretion pathways with intact liposomes (e.g. phospholipids, metallic radionuclides) and even show significant tumour uptake by themselves (e.g. some radionuclides). Finally, we describe preclinical and clinical studies using radiolabelled liposomes and discuss their impact in supporting liposomal drug development and clinical translation in several diseases, including personalised nanomedicine approaches.
Collapse
Affiliation(s)
- Francis Man
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Peter J Gawne
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, United Kingdom; London Centre for Nanotechnology, King's College London, Strand Campus, London WC2R 2LS, United Kingdom.
| |
Collapse
|
36
|
Rukavina Z, Šegvić Klarić M, Filipović-Grčić J, Lovrić J, Vanić Ž. Azithromycin-loaded liposomes for enhanced topical treatment of methicillin-resistant Staphyloccocus aureus (MRSA) infections. Int J Pharm 2018; 553:109-119. [PMID: 30312749 DOI: 10.1016/j.ijpharm.2018.10.024] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/22/2018] [Accepted: 10/08/2018] [Indexed: 12/21/2022]
Abstract
Antibiotic delivery via liposomal encapsulation represents a promising approach for the efficient topical treatment of skin infections. The present study aimed to investigate the potential of using different types of azithromycin (AZT)-loaded liposomes to locally treat skin infections caused by methicillin-resistant Staphylococcus aureus (MRSA) strains. Conventional liposomes (CLs), deformable liposomes (DLs), propylene glycol-containing liposomes (PGLs) and cationic liposomes (CATLs) encapsulating AZT were prepared, and their physical characteristics, drug release profiles, ex vivo skin penetration/deposition abilities, in vitro anti-MRSA activities (planktonic bacteria and biofilm) and cell biocompatibilities were assessed. The (phospho)lipid composition and presence of surfactant or propylene glycol affected the physical characteristics of the liposomes, the release profile of AZT, its deposition inside the skin, as well as in vitro antibacterial efficacy and tolerability with the skin cells. All the liposomes retained AZT inside the skin more efficiently than did the control and were biocompatible with keratinocytes and fibroblasts. CATLs, DLs and PGLs efficiently inhibited MRSA strain growth and were superior to free AZT in preventing biofilm formation, exhibiting minimal inhibitory concentrations and minimal biofilm inhibitory concentrations up to 32-fold lower than those of AZT solution, thus confirming their potential for improved topical treatment of MRSA-caused skin infections.
Collapse
Affiliation(s)
- Zora Rukavina
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, A. Kovačića 1, 10000 Zagreb, Croatia.
| | - Maja Šegvić Klarić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Microbiology, A. Kovačića 1, 10000 Zagreb, Croatia.
| | - Jelena Filipović-Grčić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, A. Kovačića 1, 10000 Zagreb, Croatia.
| | - Jasmina Lovrić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, A. Kovačića 1, 10000 Zagreb, Croatia.
| | - Željka Vanić
- University of Zagreb, Faculty of Pharmacy and Biochemistry, Department of Pharmaceutical Technology, A. Kovačića 1, 10000 Zagreb, Croatia.
| |
Collapse
|
37
|
Narayan R, Nayak UY, Raichur AM, Garg S. Mesoporous Silica Nanoparticles: A Comprehensive Review on Synthesis and Recent Advances. Pharmaceutics 2018; 10:E118. [PMID: 30082647 PMCID: PMC6160987 DOI: 10.3390/pharmaceutics10030118] [Citation(s) in RCA: 438] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/28/2018] [Accepted: 07/31/2018] [Indexed: 12/18/2022] Open
Abstract
Recent advancements in drug delivery technologies utilizing a variety of carriers have resulted in a path-breaking revolution in the approach towards diagnosis and therapy alike in the current times. Need for materials with high thermal, chemical and mechanical properties have led to the development of mesoporous silica nanoparticles (MSNs). These ordered porous materials have garnered immense attention as drug carriers owing to their distinctive features over the others. They can be synthesized using a relatively simple process, thus making it cost effective. Moreover, by controlling the parameters during the synthesis; the morphology, pore size and volume and particle size can be transformed accordingly. Over the last few years, a rapid increase in research on MSNs as drug carriers for the treatment of various diseases has been observed indicating its potential benefits in drug delivery. Their widespread application for the loading of small molecules as well as macromolecules such as proteins, siRNA and so forth, has made it a versatile carrier. In the recent times, researchers have sorted to several modifications in the framework of MSNs to explore its potential in drug resistant chemotherapy, antimicrobial therapy. In this review, we have discussed the synthesis of these multitalented nanoparticles and the factors influencing the size and morphology of this wonder carrier. The second part of this review emphasizes on the applications and the advances made in the MSNs to broaden the spectrum of its use especially in the field of biomedicine. We have also touched upon the lacunae in the thorough understanding of its interaction with a biological system which poses a major hurdle in the passage of this carrier to the clinical level. In the final part of this review, we have discussed some of the major patents filed in the field of MSNs for therapeutic purpose.
Collapse
Affiliation(s)
- Reema Narayan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences,Manipal Academy of Higher Education, Manipal 576104, India.
| | - Usha Y Nayak
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences,Manipal Academy of Higher Education, Manipal 576104, India.
| | - Ashok M Raichur
- Department of Materials Engineering, Indian Institute of Science, Bengaluru 560012, India.
| | - Sanjay Garg
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
38
|
Pushparaj Selvadoss P, Nellore J, Balaraman Ravindrran M, Sekar U. Novel pyochelin-based PEGylated liposomes for enhanced delivery of antibiotics against resistant clinical isolates of Pseudomonas aeruginosa. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:2043-2053. [PMID: 29179607 DOI: 10.1080/21691401.2017.1408119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Pseudomonas aeruginosa is a problematic human pathogen resistant to almost all available antibiotics. The important prerequisite for these drugs to target this bacterium is an efficient delivery system. Siderophore-mediated drug delivery system is a promising approach to carry out antibiotics to the cells. Pyochelin, a siderophore of P. aeruginosa, was successfully synthesized in a five-step procedure. PEGylated liposomal pyochelin-antibiotic (L-Pch-Ab) carrier was fabricated by thin-film hydration method. L-Pch-Ab had an average size of 90.31 ± 0.11 nm holding a negative zeta potential at -54.12 ± 0.03 mV (PDI <2). The MIC determined by broth dilution method against three clinical strains isolated from burn wounds showed that L-Pch-Ab significantly reduced (≤16 µg/ml) the MIC values than those of free antibiotics. In the time kill assay, L-Pch-Ab was bactericidal against all strains at most time intervals at 2 × and 4 × MIC up to 24 h. TEM observations revealed that L-Pch-Ab was actively taken up by P. aeruginosa and exhibited membrane deformation within 2 h. Developed L-Pch-Ab fused intimately with the outer membrane of MDRPa and exhibited effective antibacterial activity than free Ab. Furthermore, L-Pch-Ab kills MDRPa within infected HaCaT keratinocytes without any cytotoxic effects at 4× MIC concentrations after 72 h. Thus, the specific targeting of L-Pch-Ab with its higher efficacy to deliver drug by limiting the toxicity will be a novel approach to fight infections caused by P. aeruginosa.
Collapse
Affiliation(s)
| | - Jayshree Nellore
- b Department of Biotechnology , Sathyabama University , Chennai , India
| | | | - Uma Sekar
- d Department of Microbiology , Sri Ramachandra University , Chennai , India
| |
Collapse
|
39
|
Yang Z, Tian L, Liu J, Huang G. Construction and evaluation in vitro and in vivo of tedizolid phosphate loaded cationic liposomes. J Liposome Res 2017; 28:322-330. [PMID: 28920493 DOI: 10.1080/08982104.2017.1380665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
First, the SA-TDZA-Lips were prepared by reverse-phase evaporation method. Then, the drug release behaviour was evaluated by dynamic membrane dialysis in vitro and the preliminary safety was evaluated by haemolysis method. Finally, with tedizolid phosphate injection (TDZA-Inj) and tedizolid phosphate loaded liposomes (TDZA-Lips) as the control groups, the pharmacokinetic characteristic and tissues distribution of SA-TDZA-Lips were evaluated after intravenous injection. As a result, the stearylamine modified tedizolid phosphate liposomal delivery system was constructed successfully and the particle size was 194.9 ± 2.93 nm. The encapsulation efficiency (EE) was 53.52 ± 2.18%. The in vitro release of SA-TDZA-Lips was in accordance with Weibull equation. And there was no haemolysis happened, which indicated good preliminary safety for injection. The results of pharmacokinetics showed that the t1/2β increased by 0.74 times and 0.51 times higher than that of TDZA-Inj group and TDZA-Lips group, respectively. The MRT of SA-TDZA-Lips was 1.30 and 1.09 times higher than that of TDZA-Inj group and TDZA-Lips group, respectively. The AUC was 2.40 times and 0.23 times higher than that of TDZA-Inj group and TDZA-Lips group, respectively. The tissue distribution results showed that the relative uptake rate (Re) of TDZA in the lung was 1.527, which indicated the targeting. In conclusion, the SA-TDZA-Lips prepared in this study had several advantages like positive charge, strong cell affinity, prolonged circulation time in vivo, sustained release effect, and increased drug concentration in lungs. All advantages above provided significant clinical value of application for the treatment of bacterial pneumonia with tedizolid phosphate.
Collapse
Affiliation(s)
- Zhenlei Yang
- a School of Pharmaceutical Sciences , Shandong University , Jinan , PR China
| | - Liu Tian
- a School of Pharmaceutical Sciences , Shandong University , Jinan , PR China
| | - Jingjing Liu
- a School of Pharmaceutical Sciences , Shandong University , Jinan , PR China
| | - Guihua Huang
- a School of Pharmaceutical Sciences , Shandong University , Jinan , PR China
| |
Collapse
|
40
|
Alhariri M, Majrashi MA, Bahkali AH, Almajed FS, Azghani AO, Khiyami MA, Alyamani EJ, Aljohani SM, Halwani MA. Efficacy of neutral and negatively charged liposome-loaded gentamicin on planktonic bacteria and biofilm communities. Int J Nanomedicine 2017; 12:6949-6961. [PMID: 29075113 PMCID: PMC5609801 DOI: 10.2147/ijn.s141709] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We investigated the efficacy of liposomal gentamicin formulations of different surface charges against Pseudomonas aeruginosa and Klebsiella oxytoca. The liposomal gentamicin formulations were prepared by the dehydration-rehydration method, and their sizes and zeta potential were measured. Gentamicin encapsulation efficiency inside the liposomal formulations was determined by microbiologic assay, and stability of the formulations in biologic fluid was evaluated for a period of 48 h. The minimum inhibitory concentration and the minimum bactericidal concentration were determined, and the in vitro time kill studies of the free form of gentamicin and liposomal gentamicin formulations were performed. The activities of liposomal gentamicin in preventing and reducing biofilm-forming P. aeruginosa and K. oxytoca were compared to those of free antibiotic. The sizes of the liposomal formulations ranged from 625 to 806.6 nm in diameter, with the zeta potential ranging from -0.22 to -31.7 mV. Gentamicin encapsulation efficiency inside the liposomal formulation ranged from 1.8% to 43.6%. The liposomes retained >60% of their gentamicin content during the 48 h time period. The minimum inhibitory concentration of neutral formulation was lower than that of free gentamicin (0.25 versus 1 mg/L for P. aeruginosa and 0.5 versus 1 mg/L for K. oxytoca). The negatively charged formulation exhibited the same bacteriostatic concentration as that of free gentamicin. The minimum bactericidal concentration of neutral liposomes on planktonic bacterial culture was twofold lower than that of free gentamicin, whereas the negatively charged formulations were comparable to free gentamicin. The killing time curve values for the neutral negatively charged formulation against planktonic P. aeruginosa and K. oxytoca were better than those of free gentamicin. Furthermore, liposomal formulations prevent the biofilm-formation ability of these strains better than free gentamicin. In summary, liposomal formulations could be an effective lipid nanoparticle to combat acute infections where planktonic bacteria are predominant.
Collapse
Affiliation(s)
- Moayad Alhariri
- Nanomedicine Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences
| | - Majed A Majrashi
- National Centre for Biotechnology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST)
| | - Ali H Bahkali
- Botany and Microbiology Department, College of Science, King Saud University
| | - Faisal S Almajed
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Ali O Azghani
- Department of Biology, The University of Texas at Tyler, Tyler, TX, USA
| | - Mohammad A Khiyami
- National Centre for Biotechnology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST)
| | - Essam J Alyamani
- National Centre for Biotechnology, Life Sciences and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST)
| | - Sameera M Aljohani
- College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Majed A Halwani
- Nanomedicine Department, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences
| |
Collapse
|
41
|
Scomparin A, Florindo HF, Tiram G, Ferguson EL, Satchi-Fainaro R. Two-step polymer- and liposome-enzyme prodrug therapies for cancer: PDEPT and PELT concepts and future perspectives. Adv Drug Deliv Rev 2017; 118:52-64. [PMID: 28916497 DOI: 10.1016/j.addr.2017.09.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/17/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022]
Abstract
Polymer-directed enzyme prodrug therapy (PDEPT) and polymer enzyme liposome therapy (PELT) are two-step therapies developed to provide anticancer drugs site-selective intratumoral accumulation and release. Nanomedicines, such as polymer-drug conjugates and liposomal drugs, accumulate in the tumor site due to extravasation-dependent mechanism (enhanced permeability and retention - EPR - effect), and further need to cross the cellular membrane and release their payload in the intracellular compartment. The subsequent administration of a polymer-enzyme conjugate able to accumulate in the tumor tissue and to trigger the extracellular release of the active drug showed promising preclinical results. The development of polymer-enzyme, polymer-drug conjugates and liposomal drugs had undergone a vast advancement over the past decades. Several examples of enzyme mimics for in vivo therapy can be found in the literature. Moreover, polymer therapeutics often present an enzyme-sensitive mechanism of drug release. These nanomedicines can thus be optimal substrates for PDEPT and this review aims to provide new insights and stimuli toward the future perspectives of this promising combination.
Collapse
Affiliation(s)
- Anna Scomparin
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel
| | - Elaine L Ferguson
- Advanced Therapies Group, Oral and Biomedical Sciences, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Heath Park, Cardiff CF14 4XY, UK
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler School of Medicine, Room 607, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
42
|
Cui H, Yuan L, Li W, Lin L. Edible film incorporated with chitosan andArtemisia annuaoil nanoliposomes for inactivation ofEscherichia coliO157:H7 on cherry tomato. Int J Food Sci Technol 2016. [DOI: 10.1111/ijfs.13322] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Haiying Cui
- School of Food and Biological Engineering; Jiangsu University; Zhenjiang 212013 China
| | - Lu Yuan
- School of Food and Biological Engineering; Jiangsu University; Zhenjiang 212013 China
| | - Wei Li
- School of Food and Biological Engineering; Jiangsu University; Zhenjiang 212013 China
| | - Lin Lin
- School of Food and Biological Engineering; Jiangsu University; Zhenjiang 212013 China
| |
Collapse
|
43
|
Li Y, Tang C, Zhang E, Yang L. Colistin-entrapped liposomes driven by the electrostatic interaction: Mechanism of drug loading and in vivo characterization. Int J Pharm 2016; 515:20-29. [PMID: 27713028 DOI: 10.1016/j.ijpharm.2016.10.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/16/2016] [Accepted: 10/03/2016] [Indexed: 11/19/2022]
Abstract
The potential in vivo application of liposome for polycationic colistin has been hindered by the poor entrapment efficiency (EE) due to their phospholipid membrane permeability. The objective of this study is to investigate the loading mechanism and validity of applying electrostatic attraction for the colistin entrapment and delivery in liposomes. Anionic lipids with various structures were used for colistin entrapment, and the properties of resulting liposomes (i.e. zeta-potential, EE and release rate) were highly dependent on the structure of anionic lipids. Based on consideration of intermolecular interactions, the retention of electrostatically entrapped colistin is essentially determined by the balance of interfacial hydrophobic attraction and electrostatic repulsion. The liposomal colistin showed the reduced bacterial killing rate, but did not compromise the in vitro antibacterial activity. Specially, the PEGylated liposomal colistin of sodium cholesteryl sulfate (Chol-SO4-) showed the best drug retention, resulting in the significantly increased maximum-tolerated dose, prolonged blood circulation and decreased colistin distribution in kidney after intravenous administration in mice. These results highlight the potential utility of electrostatically entrapped liposome for polycationic colistin delivery.
Collapse
Affiliation(s)
- Yang Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Chengcheng Tang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Enbo Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Li Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
44
|
Zahra MJ, Hamed H, Mohammad RY, Nosratollah Z, Akbarzadeh A, Morteza M. Evaluation and study of antimicrobial activity of nanoliposomal meropenem against Pseudomonas aeruginosa isolates. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2016; 45:975-980. [DOI: 10.1080/21691401.2016.1198362] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Momeni-Javid Zahra
- Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Hamishekar Hamed
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rahmati-Yamchi Mohammad
- Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zarghami Nosratollah
- Department of Medical Biotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Science, Tabriz, Iran
| | - Abolfazl Akbarzadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milani Morteza
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
45
|
Current Trends in Development of Liposomes for Targeting Bacterial Biofilms. Pharmaceutics 2016; 8:pharmaceutics8020018. [PMID: 27231933 PMCID: PMC4932481 DOI: 10.3390/pharmaceutics8020018] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/17/2016] [Accepted: 05/18/2016] [Indexed: 12/26/2022] Open
Abstract
Biofilm targeting represents a great challenge for effective antimicrobial therapy. Increased biofilm resistance, even with the elevated concentrations of very potent antimicrobial agents, often leads to failed therapeutic outcome. Application of biocompatible nanomicrobials, particularly liposomally-associated nanomicrobials, presents a promising approach for improved drug delivery to bacterial cells and biofilms. Versatile manipulations of liposomal physicochemical properties, such as the bilayer composition, membrane fluidity, size, surface charge and coating, enable development of liposomes with desired pharmacokinetic and pharmacodynamic profiles. This review attempts to provide an unbiased overview of investigations of liposomes destined to treat bacterial biofilms. Different strategies including the recent advancements in liposomal design aiming at eradication of existing biofilms and prevention of biofilm formation, as well as respective limitations, are discussed in more details.
Collapse
|
46
|
Current applications of nanoparticles in infectious diseases. J Control Release 2016; 224:86-102. [PMID: 26772877 DOI: 10.1016/j.jconrel.2016.01.008] [Citation(s) in RCA: 254] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 02/06/2023]
Abstract
For decades infections have been treated easily with drugs. However, in the 21st century, they may become lethal again owing to the development of antimicrobial resistance. Pathogens can become resistant by means of different mechanisms, such as increasing the time they spend in the intracellular environment, where drugs are unable to reach therapeutic levels. Moreover, drugs are also subject to certain problems that decrease their efficacy. This requires the use of high doses, and frequent administrations must be implemented, causing adverse side effects or toxicity. The use of nanoparticle systems can help to overcome such problems and increase drug efficacy. Accordingly, there is considerable current interest in their use as antimicrobial agents against different pathogens like bacteria, virus, fungi or parasites, multidrug-resistant strains and biofilms; as targeting vectors towards specific tissues; as vaccines and as theranostic systems. This review begins with an overview of the different types and characteristics of nanoparticles used to deliver drugs to the target, followed by a review of current research and clinical trials addressing the use of nanoparticles within the field of infectious diseases.
Collapse
|
47
|
Serrano Figueroa LO, Pitts B, Uchida M, Richards AM. Vesicle self-assembly of amphiphilic siderophores produced by bacterial isolates from Soap Lake, Washington. CAN J CHEM 2016. [DOI: 10.1139/cjc-2015-0173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Soap Lake, located in Washington State, is a meromictic soda lake that was the subject of a prior National Science Foundation funded Microbial Observatory. Several organisms inhabiting this lake have been identified as producers of siderophores that are unique in structure. Two isolates found to be of the species Halomonas, SL01 and SL28, were found to produce suites of amphiphilic siderophores consisting of a peptidic head-group, which binds iron appended to fatty acid moieties of various lengths. The ability for siderophores to self-assemble into vesicles was determined for three suites of amphiphilic siderophores of unique structure (two from SL01 and one from SL28). These siderophores resemble the amphiphilic aquachelin siderophores produced by Halomonas aquamarina strain DS40M3, a marine bacterium. Vesicle self-assembly studies were performed by dynamic light scattering and epifluorescence microscopy. The addition of ferric iron (Fe3+) at different equivalents, where an equivalence of iron is defined as equal to the molarity of the siderophore, demonstrated vesicle formation. This was suggested by both dynamic light scattering and epifluorescence microscopy. Bacteria thriving under saline and alkaline conditions are capable of producing unique siderophores that self-assemble in micelles and vesicles due to ferric iron chelation.
Collapse
Affiliation(s)
- Luis O’mar Serrano Figueroa
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
| | - Betsey Pitts
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
| | - Masaki Uchida
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, MT 59717, USA
| | - Abigail M. Richards
- Center for Biofilm Engineering, Montana State University, Bozeman, MT 59717, USA
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT 59717, USA
| |
Collapse
|
48
|
Wendel SO, Menon S, Alshetaiwi H, Shrestha TB, Chlebanowski L, Hsu WW, Bossmann SH, Narayanan S, Troyer DL. Cell Based Drug Delivery: Micrococcus luteus Loaded Neutrophils as Chlorhexidine Delivery Vehicles in a Mouse Model of Liver Abscesses in Cattle. PLoS One 2015; 10:e0128144. [PMID: 26011247 PMCID: PMC4444037 DOI: 10.1371/journal.pone.0128144] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 04/22/2015] [Indexed: 01/13/2023] Open
Abstract
The recent WHO report on antibiotic resistances shows a dramatic increase of microbial resistance against antibiotics. With only a few new antibiotics in the pipeline, a different drug delivery approach is urgently needed. We have obtained evidence demonstrating the effectiveness of a cell based drug delivery system that utilizes the innate immune system as targeting carrier for antibacterial drugs. In this study we show the efficient loading of neutrophil granulocytes with chlorhexidine and the complete killing of E. coli as well as Fusobacterium necrophorum in in-vitro studies. Fusobacterium necrophorum causes hepatic abscesses in cattle fed high grain diets. We also show in a mouse model that this delivery system targets infections of F. necrophorum in the liver and reduces the bacterial burden by an order of magnitude from approximately 2•106 to 1•105.
Collapse
Affiliation(s)
- Sebastian O. Wendel
- Department of Chemical Engineering, Kansas State University, Manhattan, Kansas, United States of America
| | - Sailesh Menon
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Hamad Alshetaiwi
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States of America
- University of Ha'il, Ha'il, Saudi Arabia
| | - Tej B. Shrestha
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Lauren Chlebanowski
- Department of Chemistry, Augustana College, Rock Island, Illinois, United States of America
| | - Wei-Wen Hsu
- Department of Statistics, Kansas State University, Manhattan, Kansas, United States of America
| | - Stefan H. Bossmann
- Department of Chemistry, Kansas State University, Manhattan, Kansas, United States of America
| | - Sanjeev Narayanan
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Deryl L. Troyer
- Department of Anatomy and Physiology, Kansas State University, Manhattan, Kansas, United States of America
| |
Collapse
|
49
|
Zhou QT, Leung SSY, Tang P, Parumasivam T, Loh ZH, Chan HK. Inhaled formulations and pulmonary drug delivery systems for respiratory infections. Adv Drug Deliv Rev 2015; 85:83-99. [PMID: 25451137 DOI: 10.1016/j.addr.2014.10.022] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/15/2014] [Accepted: 10/18/2014] [Indexed: 11/16/2022]
Abstract
Respiratory infections represent a major global health problem. They are often treated by parenteral administrations of antimicrobials. Unfortunately, systemic therapies of high-dose antimicrobials can lead to severe adverse effects and this calls for a need to develop inhaled formulations that enable targeted drug delivery to the airways with minimal systemic drug exposure. Recent technological advances facilitate the development of inhaled anti-microbial therapies. The newer mesh nebulisers have achieved minimal drug residue, higher aerosolisation efficiencies and rapid administration compared to traditional jet nebulisers. Novel particle engineering and intelligent device design also make dry powder inhalers appealing for the delivery of high-dose antibiotics. In view of the fact that no new antibiotic entities against multi-drug resistant bacteria have come close to commercialisation, advanced formulation strategies are in high demand for combating respiratory 'super bugs'.
Collapse
Affiliation(s)
- Qi Tony Zhou
- Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sharon Shui Yee Leung
- Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Patricia Tang
- Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Thaigarajan Parumasivam
- Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia
| | - Zhi Hui Loh
- GEA-NUS Pharmaceutical Processing Research Laboratory, Department of Pharmacy, National University of Singapore, 18 Science Drive 4, Singapore 117543, Singapore
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, Faculty of Pharmacy, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
50
|
Nanotechnology approaches for antibacterial drug delivery: Preparation and microbiological evaluation of fusogenic liposomes carrying fusidic acid. Int J Antimicrob Agents 2015; 45:622-6. [PMID: 25816979 DOI: 10.1016/j.ijantimicag.2015.01.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2014] [Revised: 01/17/2015] [Accepted: 01/21/2015] [Indexed: 11/24/2022]
Abstract
Many antibacterial drugs have some difficulty passing through the bacterial cell membrane, especially if they have a high molecular weight or large spatial structure. Consequently, intrinsic resistance is shown by some bacterial strains. Reduced cell membrane permeability is one of the mechanisms of resistance known for fusidic acid (FUS), a bacteriostatic steroidal compound with activity limited to Gram-positive bacteria. Moreover, the lipophilic character of FUS has been shown to cause drug retention inside the bilayers of cell membranes, preventing its diffusion towards target sites inside the cytoplasm. Targeting antimicrobial agents by means of liposomes may be a valid strategy in the treatment of infections refractory to conventional routes of antimicrobial treatment. On this basis, loading of FUS in fusogenic liposomes (FLs) was planned in this study. Fusogenic small unilamellar vesicles loaded with FUS were produced to evaluate their influence on improving the cell penetration and antibacterial activity of the antibiotic. The produced carriers were technologically characterised and were subjected to an in vitro microbiological assay against several strains of Gram-negative and Gram-positive bacteria. The experimental results showed that encapsulating FUS in a liposomal carrier can improve antimicrobial efficacy and reduce the effective concentration required, probably through putative mechanisms of increased diffusion through the bacterial cell membrane. In fact, whilst free FUS was active only on the tested Gram-positive strains, incubation of FUS-loaded FLs exhibited growth inhibitory activity both against Gram-positive and Gram-negative strains. The lowest MICs were obtained against Staphylococcus epidermidis (≤0.15 μg/mL) and Acinetobacter baumannii (37.5 μg/mL) clinical strains.
Collapse
|