1
|
Kharwade R, Kazi M, Mahajan N, Badole P, More S, Kayali A, Noushad Javed M, Kaleem M. Mannosylated PAMAM G2 dendrimers mediated rate programmed delivery of efavirenz target HIV viral latency at reservoirs. Saudi Pharm J 2024; 32:102154. [PMID: 39282004 PMCID: PMC11399684 DOI: 10.1016/j.jsps.2024.102154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 08/07/2024] [Indexed: 09/18/2024] Open
Abstract
In this current research, we conceptualized a novel nanotechnology-enabled synthesis approach of targeting HIV-harboring tissues via second-generation (G2) polyamidoamine (PAMAM) mannosylated (MPG2) dendrimers for programmed delivery of anti-HIV drugs efavirenz (EFV) and ritonavir (RTV). Briefly, here mannose served purpose of ligand in this EFV and RTV-loaded PAMAM G2 dendrimers, synthesized by divergent techniques, denoted as MPG2ER. The developed nanocarriers were characterized by different analytical tools FTIR, NMR, zeta potential, particle size, and surface morphology. The results of confocal microscopy showed substantial alterations in the morphology of H9 cells, favored by relatively higher drug uptake through the MPG2ER. Interestingly, the drug uptake study and cytotoxicity assay of MPG2ER demonstrated that it showed no significant toxicity up to 12.5 µM. A typical flow cytometry histogram also revealed that MPG2ER efficiently internalized both drugs, with an increase in drug uptake of up to 81.2 %. It also enhanced the plasma pharmacokinetics of EFV, with Cmax7.68 μg/ml, AUC of 149.19 (μg/ml) * hr, and MRT of 26.87 hrs. Subsequently, tissue pharmacokinetics further evidence that MPG2ER accumulated more in distant Human immunodeficiency virus (HIV) reservoir tissues, such as the lymph nodes and spleen, but without exhibiting significant toxicity. Abovementioned compelling evidences strongly favored translational roles of MPG2 as a potential therapeutic strategy in the clinical eradication of HIV from viral reservoir tissue.
Collapse
Affiliation(s)
- Rohini Kharwade
- Department of Pharmaceutics, Dadasaheb Balpande College of Pharmacy, Besa, Nagpur, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, MS, India
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Nilesh Mahajan
- Department of Pharmaceutics, Dadasaheb Balpande College of Pharmacy, Besa, Nagpur, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, MS, India
| | - Payal Badole
- Department of Pharmaceutics, Dadasaheb Balpande College of Pharmacy, Besa, Nagpur, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, MS, India
| | - Sachin More
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra 440037, India
| | - Asaad Kayali
- Department of Biomedical Sciences, College of Health Science, Abu Dhabi University, Abu Dhabi P.O. Box 59911, United Arab Emirates
| | - Md Noushad Javed
- NationNanotechnology Center of Excellence, College of Engineering and Computer Science, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Mohammed Kaleem
- Department of Pharmacology, Dadasaheb Balpande College of Pharmacy, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur, Maharashtra 440037, India
| |
Collapse
|
2
|
Yang J, Shi C, Cheng Y, Zhu Y, Yang X, Liang Y, Liang H, Lin Q, Li M, Xun J, Liu J, Yin C, Qi J, Zhu H. Effective in vivo reactivation of HIV-1 latency reservoir via oral administration of EK-16A-SNEDDS. Eur J Pharm Biopharm 2024; 201:114353. [PMID: 38885911 DOI: 10.1016/j.ejpb.2024.114353] [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: 03/22/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
The latent reservoir of human immunodeficiency virus (HIV) is a major obstacle in the treatment of acquired immune deficiency syndrome (AIDS). The "shock and kill" strategy has emerged as a promising approach for clearing HIV latent reservoirs. However, current latency-reversing agents (LRAs) have limitations in effectively and safely activating the latent virus and reducing the HIV latent reservoirs in clinical practice. Previously, EK-16A was extracted from Euphorbia kansui, which had the effect of interfering with the HIV-1 latent reservoir and inhibiting HIV-1 entry. Nevertheless, there is no suitable and efficient EK-16A oral formulation for in vivo delivery and clinical use. In this study, an oral EK-16A self-nanoemulsifying drug delivery system (EK-16A-SNEDDS) was proposed to "shock" the HIV-1 latent reservoir. This system aims to enhance the bioavailability and delivery of EK-16A to various organs. The composition of EK-16A-SNEDDS was optimized through self-emulsifying grading and ternary phase diagram tests. Cell models, pharmacokinetic experiments, and pharmacodynamics in HIV-1 latent cell transplant animal models suggested that EK-16A-SNEDDS could be absorbed by the gastrointestinal tract and enter the blood circulation after oral administration, thereby reaching various organs to activate latent HIV-1. The prepared EK-16A-SNEDDS demonstrated safety and efficacy, exhibited high clinical experimental potential, and may be a promising oral preparation for eliminating HIV-1 latent reservoirs.
Collapse
Affiliation(s)
- Jinlong Yang
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China; Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Chenyi Shi
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yipeng Cheng
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yuqi Zhu
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xinyi Yang
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China; Yiwu Research Institute of Fudan University, Yiwu 322000, China
| | - Yue Liang
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Huitong Liang
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Qinru Lin
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Min Li
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jingna Xun
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jianping Liu
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Jianping Qi
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Huanzhang Zhu
- State Key Laboratory of Genetic Engineering and Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
3
|
Rojekar S, Gholap AD, Togre N, Bhoj P, Haeck C, Hatvate N, Singh N, Vitore J, Dhoble S, Kashid S, Patravale V. Current status of mannose receptor-targeted drug delivery for improved anti-HIV therapy. J Control Release 2024; 372:494-521. [PMID: 38849091 DOI: 10.1016/j.jconrel.2024.06.002] [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: 03/12/2024] [Revised: 05/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
In the pursuit of achieving better therapeutic outcomes in the treatment of HIV, innovative drug delivery strategies have been extensively explored. Mannose receptors, which are primarily found on macrophages and dendritic cells, offer promising targets for drug delivery due to their involvement in HIV pathogenesis. This review article comprehensively evaluates recent drug delivery system advancements targeting the mannose receptor. We have systematically described recent developments in creating and utilizing drug delivery platforms, including nanoparticles, liposomes, micelles, noisomes, dendrimers, and other nanocarrier systems targeted at the mannose receptor. These strategies aim to enhance drug delivery specificity, bioavailability, and therapeutic efficacy while decreasing off-target effects and systemic toxicity. Furthermore, the article delves into how mannose receptors and HIV interact, highlighting the potential for exploiting this interaction to enhance drug delivery to infected cells. The review covers essential topics, such as the rational design of nanocarriers for mannose receptor recognition, the impact of physicochemical properties on drug delivery performance, and how targeted delivery affects the pharmacokinetics and pharmacodynamics of anti-HIV agents. The challenges of these novel strategies, including immunogenicity, stability, and scalability, and future research directions in this rapidly growing area are discussed. The knowledge synthesis presented in this review underscores the potential of mannose receptor-based targeted drug delivery as a promising avenue for advancing HIV treatment. By leveraging the unique properties of mannose receptors, researchers can design drug delivery systems that cater to individual needs, overcome existing limitations, and create more effective and patient-friendly treatments in the ongoing fight against HIV/AIDS.
Collapse
Affiliation(s)
- Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Namdev Togre
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Priyanka Bhoj
- Department of Pathology, Lewis Katz School of Medicine at Temple University, Philadelphia, USA
| | - Clement Haeck
- Population Council, , Center for Biomedical Research, 1230 York Avenue, New York, NY 10065, USA
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nidhi Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India
| | - Jyotsna Vitore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Sagar Dhoble
- Department of Pharmacology and Toxicology, R. K. Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Snehal Kashid
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gujarat 382355, India
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India.
| |
Collapse
|
4
|
Gaikwad SY, Tyagi S, Seniya C, More A, Chandane-Tak M, Kumar S, Mukherjee A. A nanoemulsified formulation of dolutegravir and epigallocatechin gallate inhibits HIV-1 replication in cellular models. FEBS Lett 2024; 598:1919-1936. [PMID: 38789398 DOI: 10.1002/1873-3468.14936] [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: 04/05/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024]
Abstract
Nanotechnology offers promising avenues for enhancing drug delivery systems, particularly in HIV-1 treatment. This study investigates a nanoemulsified formulation combining epigallocatechin gallate (EGCG) with dolutegravir (DTG) for managing HIV-1 infection. The combinatorial interaction between EGCG and DTG was explored through cellular, enzymatic, and molecular studies. In vitro assays demonstrated the potential of a dual drug-loaded nanoemulsion, NE-DTG-EGCG, in inhibiting HIV-1 replication, with EGCG serving as a supplementary treatment containing DTG. In silico molecular interaction studies highlighted EGCG's multifaceted inhibitory potential against HIV-1 integrase and reverse transcriptase enzymes. Further investigations are needed to validate the formulation's efficacy across diverse contexts. Overall, by integrating nanotechnology into drug delivery systems, this study represents a significant advancement in managing HIV-1 infection.
Collapse
Affiliation(s)
- Shraddha Y Gaikwad
- Division of Virology, ICMR-National AIDS Research Institute, Pune, India
| | - Shivani Tyagi
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, India
| | - Chandrabhan Seniya
- School of Biosciences, Engineering and Technology, VIT Bhopal University, India
| | - Ashwini More
- Division of Virology, ICMR-National AIDS Research Institute, Pune, India
| | | | - Shobhit Kumar
- School of Biosciences, Engineering and Technology, VIT Bhopal University, India
| | - Anupam Mukherjee
- Division of Virology, ICMR-National AIDS Research Institute, Pune, India
| |
Collapse
|
5
|
Kumar S, Taumar D, Gaikwad S, More A, Nema V, Mukherjee A. Antiretroviral action of Rosemary oil-based atazanavir formulation and the role of self-nanoemulsifying drug delivery system in the management of HIV-1 infection. Drug Deliv Transl Res 2024; 14:1888-1908. [PMID: 38161197 DOI: 10.1007/s13346-023-01492-8] [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] [Accepted: 12/06/2023] [Indexed: 01/03/2024]
Abstract
Atazanavir or ATV is an FDA-approved, HIV-1 protease inhibitor that belongs to the azapeptide group. Over time, it has been observed that ATV can cause multiple adverse side effects in the form of liver diseases including elevations in serum aminotransferase, indirect hyper-bilirubinemia, and idiosyncratic acute liver injury aggravating the underlying chronic viral hepatitis. Hence, there is an incessant need to explore the safe and efficacious method of delivering ATV in a controlled manner that may reduce the proportion of its idiosyncratic reactions in patients who are on antiretroviral therapy for years. In this study, we assessed ATV formulation along with Rosemary oil to enhance the anti-HIV-1 activity and its controlled delivery through self-nanoemulsifying drug delivery system or SNEDDS to enhance its oral bioavailability. While the designing, development, and characterization of ATV-SNEDDS were addressed through various evaluation parameters and pharmacokinetic-based studies, in vitro cell-based experiments assured the safety and efficacy of the designed ATV formulation. The study discovered the potential of ATV-SNEDDS to inhibit HIV-1 infection at a lower concentration as compared to its pure counterpart. Simultaneously, we could also demonstrate the ATV and Rosemary oil providing leads for designing and developing such formulations for the management of HIV-1 infections with the alleviation in the risk of adverse reactions.
Collapse
Affiliation(s)
- Shobhit Kumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology (MIET), NH-58, Delhi-Roorkee Highway, Meerut, 250005, Uttar Pradesh, India
| | - Dhananjay Taumar
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology (MIET), NH-58, Delhi-Roorkee Highway, Meerut, 250005, Uttar Pradesh, India
| | - Shraddha Gaikwad
- Division of Virology, ICMR-National AIDS Research Institute, Ministry of Health & Family Welfare, Plot No. 73, 'G' Block, MIDC, Bhosari, Pune, 411026, Maharashtra, India
| | - Ashwini More
- Division of Virology, ICMR-National AIDS Research Institute, Ministry of Health & Family Welfare, Plot No. 73, 'G' Block, MIDC, Bhosari, Pune, 411026, Maharashtra, India
| | - Vijay Nema
- Division of Virology, ICMR-National AIDS Research Institute, Ministry of Health & Family Welfare, Plot No. 73, 'G' Block, MIDC, Bhosari, Pune, 411026, Maharashtra, India
| | - Anupam Mukherjee
- Division of Virology, ICMR-National AIDS Research Institute, Ministry of Health & Family Welfare, Plot No. 73, 'G' Block, MIDC, Bhosari, Pune, 411026, Maharashtra, India.
| |
Collapse
|
6
|
Lebrón JA, Ostos FJ, Martínez-Santa M, García-Moscoso F, López-López M, Moyá ML, Bernal E, Bachiller S, González-Ulloa G, Rodríguez-Lucena D, Lopes-Costa T, Fernández-Torres R, Ruiz-Mateos E, Pedrosa JM, Rafii-El-Idrissi Benhnia M, López-Cornejo P. Biocompatible metal-organic frameworks as promising platforms to eradicate HIV reservoirs ex vivo in people living with HIV. J Mater Chem B 2024; 12:5220-5237. [PMID: 38695162 DOI: 10.1039/d4tb00272e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
The HIV attacks the immune system provoking an infection that is considered a global health challenge. Despite antiretroviral treatments being effective in reducing the plasma viral load in the blood to undetectable levels in people living with HIV (PLWH), the disease is not cured and has become chronic. This happens because of the existence of anatomical and cellular viral reservoirs, mainly located in the lymph nodes and gastrointestinal tract, which are composed of infected CD4+ T cells with a resting memory phenotype and inaccessible to antiretroviral therapy. Herein, a new therapeutic strategy based on nanotechnology is presented. Different combinations of antiretroviral drugs (bictegravir/tenofovir/emtricitabine and nevirapine/tenofovir/emtricitabine) and toll-like receptor agonists were encapsulated into metal-organic frameworks (MOFs) PCN-224 and ZIF-8. The encapsulation efficiencies of all the drugs, as well as their release rate from the carriers, were measured. In vitro studies about the cell viability, the hemocompatibility, and the platelet aggregation of the MOFs were carried out. Epifluorescence microscopy assays confirmed the ability of ZIF-8 to target a carboxyfluorescein probe inside HeLa cell lines and PBMCs. These results pave the way for the use of these structures to eliminate latent HIV reservoirs from anatomical compartments through the activation of innate immune cells, and a higher efficacy of the triplet combinations of antiretroviral drugs.
Collapse
Affiliation(s)
- José A Lebrón
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, C/Prof. García González 1, 41012 Seville, Spain.
| | - Francisco J Ostos
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío University Hospital/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, 41013 Seville, Spain
| | - Marta Martínez-Santa
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, C/Prof. García González 1, 41012 Seville, Spain.
| | - Francisco García-Moscoso
- Department of Physical, Chemical and Natural Systems, University Pablo de Olavide, Ctra. Utrera Km. 1, 41013, Seville, Spain
| | - Manuel López-López
- Department of Chemical Engineering, Physical Chemistry and Materials Science, Campus 'El Carmen', Faculty of Experimental Sciences, University of Huelva, 21071, Huelva, Spain
| | - María L Moyá
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, C/Prof. García González 1, 41012 Seville, Spain.
| | - Eva Bernal
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, C/Prof. García González 1, 41012 Seville, Spain.
| | - Sara Bachiller
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío University Hospital/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, 41013 Seville, Spain
| | - Gabriel González-Ulloa
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío University Hospital/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, 41013 Seville, Spain
| | - David Rodríguez-Lucena
- Department of Physical, Chemical and Natural Systems, University Pablo de Olavide, Ctra. Utrera Km. 1, 41013, Seville, Spain
| | - Tania Lopes-Costa
- Department of Physical, Chemical and Natural Systems, University Pablo de Olavide, Ctra. Utrera Km. 1, 41013, Seville, Spain
| | - Rut Fernández-Torres
- Department of Analytical Chemistry, Faculty of Chemistry, University of Seville, c/Prof. García González, 1, 41012, Seville, Spain
| | - Ezequiel Ruiz-Mateos
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío University Hospital/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, 41013 Seville, Spain
| | - José M Pedrosa
- Department of Physical, Chemical and Natural Systems, University Pablo de Olavide, Ctra. Utrera Km. 1, 41013, Seville, Spain
| | - Mohammed Rafii-El-Idrissi Benhnia
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, 41009 Seville, Spain
- Institute of Biomedicine of Seville, IBiS/Virgen del Rocío University Hospital/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology and Parasitology, 41013 Seville, Spain
| | - Pilar López-Cornejo
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, C/Prof. García González 1, 41012 Seville, Spain.
| |
Collapse
|
7
|
Megantara S, Rusdin A, Budiman A, Shamsuddin S, Mohtar N, Muchtaridi M. Revolutionizing Antiviral Therapeutics: Unveiling Innovative Approaches for Enhanced Drug Efficacy. Int J Nanomedicine 2024; 19:2889-2915. [PMID: 38525012 PMCID: PMC10961067 DOI: 10.2147/ijn.s447721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Since the beginning of the coronavirus pandemic in late 2019, viral infections have become one of the top three causes of mortality worldwide. Immunization and the use of immunomodulatory drugs are effective ways to prevent and treat viral infections. However, the primary therapy for managing viral infections remains antiviral and antiretroviral medication. Unfortunately, these drugs are often limited by physicochemical constraints such as low target selectivity and poor aqueous solubility. Although several modifications have been made to enhance the physicochemical characteristics and efficacy of these drugs, there are few published studies that summarize and compare these modifications. Our review systematically synthesized and discussed antiviral drug modification reports from publications indexed in Scopus, PubMed, and Google Scholar databases. We examined various approaches that were investigated to address physicochemical issues and increase activity, including liposomes, cocrystals, solid dispersions, salt modifications, and nanoparticle drug delivery systems. We were impressed by how well each strategy addressed physicochemical issues and improved antiviral activity. In conclusion, these modifications represent a promising way to improve the physicochemical characteristics, functionality, and effectiveness of antivirals in clinical therapy.
Collapse
Affiliation(s)
- Sandra Megantara
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Research Collaboration Centre for Theranostic Radio Pharmaceuticals, National Research and Innovation Agency (BRIN), Sumedang, 45363, Indonesia
| | - Agus Rusdin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | | | - Noratiqah Mohtar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, 11800, Malaysia
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
- Research Collaboration Centre for Theranostic Radio Pharmaceuticals, National Research and Innovation Agency (BRIN), Sumedang, 45363, Indonesia
- Functional Nano Powder University Center of Excellence (FiNder U CoE), Universitas Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
8
|
Gholap AD, Kapare HS, Pagar S, Kamandar P, Bhowmik D, Vishwakarma N, Raikwar S, Garkal A, Mehta TA, Rojekar S, Hatvate N, Mohanto S. Exploring modified chitosan-based gene delivery technologies for therapeutic advancements. Int J Biol Macromol 2024; 260:129581. [PMID: 38266848 DOI: 10.1016/j.ijbiomac.2024.129581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/26/2023] [Accepted: 01/06/2024] [Indexed: 01/26/2024]
Abstract
One of the critical steps in gene therapy is the successful delivery of the genes. Immunogenicity and toxicity are major issues for viral gene delivery systems. Thus, non-viral vectors are explored. A cationic polysaccharide like chitosan could be used as a nonviral gene delivery vector owing to its significant interaction with negatively charged nucleic acid and biomembrane, providing effective cellular uptake. However, the native chitosan has issues of targetability, unpacking ability, and solubility along with poor buffer capability, hence requiring modifications for effective use in gene delivery. Modified chitosan has shown that the "proton sponge effect" involved in buffering the endosomal pH results in osmotic swelling owing to the accumulation of a greater amount of proton and chloride along with water. The major challenges include limited exploration of chitosan as a gene carrier, the availability of high-purity chitosan for toxicity reduction, and its immunogenicity. The genetic drugs are in their infancy phase and require further exploration for effective delivery of nucleic acid molecules as FDA-approved marketed formulations soon.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Harshad S Kapare
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pune 411018, Maharashtra, India
| | - Sakshi Pagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - Pallavi Kamandar
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Deblina Bhowmik
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nikhar Vishwakarma
- Department of Pharmacy, Gyan Ganga Institute of Technology and Sciences, Jabalpur 482003, Madhya Pradesh, India
| | - Sarjana Raikwar
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar 470003, Madhya Pradesh, India
| | - Atul Garkal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Tejal A Mehta
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India.
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru, Karnataka 575018, India
| |
Collapse
|
9
|
Gholap AD, Rojekar S, Kapare HS, Vishwakarma N, Raikwar S, Garkal A, Mehta TA, Jadhav H, Prajapati MK, Annapure U. Chitosan scaffolds: Expanding horizons in biomedical applications. Carbohydr Polym 2024; 323:121394. [PMID: 37940287 DOI: 10.1016/j.carbpol.2023.121394] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 11/10/2023]
Abstract
Chitosan, a natural polysaccharide from chitin, shows promise as a biomaterial for various biomedical applications due to its biocompatibility, biodegradability, antibacterial activity, and ease of modification. This review overviews "chitosan scaffolds" use in diverse biomedical applications. It emphasizes chitosan's structural and biological properties and explores fabrication methods like gelation, electrospinning, and 3D printing, which influence scaffold architecture and mechanical properties. The review focuses on chitosan scaffolds in tissue engineering and regenerative medicine, highlighting their role in bone, cartilage, skin, nerve, and vascular tissue regeneration, supporting cell adhesion, proliferation, and differentiation. Investigations into incorporating bioactive compounds, growth factors, and nanoparticles for improved therapeutic effects are discussed. The review also examines chitosan scaffolds in drug delivery systems, leveraging their prolonged release capabilities and ability to encapsulate medicines for targeted and controlled drug delivery. Moreover, it explores chitosan's antibacterial activity and potential for wound healing and infection management in biomedical contexts. Lastly, the review discusses challenges and future objectives, emphasizing the need for improved scaffold design, mechanical qualities, and understanding of interactions with host tissues. In summary, chitosan scaffolds hold significant potential in various biological applications, and this review underscores their promising role in advancing biomedical science.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Harshad S Kapare
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pune 411018, Maharashtra, India
| | - Nikhar Vishwakarma
- Department of Pharmacy, Gyan Ganga Institute of Technology and Sciences, Jabalpur 482003, Madhya Pradesh, India
| | - Sarjana Raikwar
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar 470003, Madhya Pradesh, India
| | - Atul Garkal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Tejal A Mehta
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Harsh Jadhav
- Department of Food Engineering and Technology, Institute of Chemical Technology (ICT), Mumbai 400 019, Maharashtra, India
| | - Mahendra Kumar Prajapati
- Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKM's NMIMS, Shirpur 425405, Maharashtra, India.
| | - Uday Annapure
- Institute of Chemical Technology, Marathwada Campus, Jalna 431203, Maharashtra, India; Department of Food Engineering and Technology, Institute of Chemical Technology (ICT), Mumbai 400 019, Maharashtra, India.
| |
Collapse
|
10
|
Khurana A, Allawadhi P, Singh V, Khurana I, Yadav P, Sathua KB, Allwadhi S, Banothu AK, Navik U, Bharani KK. Antimicrobial and anti-viral effects of selenium nanoparticles and selenoprotein based strategies: COVID-19 and beyond. J Drug Deliv Sci Technol 2023; 86:104663. [PMID: 37362903 PMCID: PMC10249347 DOI: 10.1016/j.jddst.2023.104663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023]
Abstract
Deficiency of selenium (Se) has been described in a significant number of COVID-19 patients having a higher incidence of mortality, which makes it a pertinent issue to be addressed clinically for effective management of the COVID-19 pandemic. Se nanoparticles (SeNPs) provide a unique option for managing the havoc caused by the COVID-19 pandemic. SeNPs possess promising anti-inflammatory and anti-fibrotic effects by virtue of their nuclear factor kappa-light-chain-stimulator of activated B cells (NFκB), mitogen-activated protein kinase (MAPKs), and transforming growth factor-beta (TGF-β) modulatory activity. In addition, SeNPs possess remarkable immunomodulatory effects, making them a suitable option for supplementation with a much lower risk of toxicity compared to their elemental counterpart. Further, SeNPs have been shown to curtail viral and microbial infections, thus, making it a novel means to halt viral growth. In addition, it can be administered in the form of aerosol spray, direct injection, or infused thin-film transdermal patches to reduce the spread of this highly contagious viral infection. Moreover, a considerable decrease in the expression of selenoprotein along with enhanced expression of IL-6 in COVID-19 suggests a potential association among selenoprotein expression and COVID-19. In this review, we highlight the unique antimicrobial and antiviral properties of SeNPs and the immunomodulatory potential of selenoproteins. We provide the rationale behind their potentially interesting properties and further exploration in the context of microbial and viral infections. Further, the importance of selenoproteins and their role in maintaining a successful immune response along with their association to Se status is summarized.
Collapse
Affiliation(s)
- Amit Khurana
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Rajendranagar, Hyderabad, 500030, PVNRTVU, Telangana, India
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Warangal, 506166, PVNRTVU, Telangana, India
| | - Prince Allawadhi
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Vishakha Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology (IIT) Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Isha Khurana
- Department of Pharmaceutical Chemistry, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, 160014, India
| | - Poonam Yadav
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Kshirod Bihari Sathua
- Department of Pharmacology, College of Pharmaceutical Sciences, Konark Marine Drive Road, Puri, 752002, Odisha, India
| | - Sachin Allwadhi
- Department of Computer Science and Engineering, University Institute of Engineering and Technology (UIET), Maharshi Dayanand University (MDU), Rohtak, 124001, Haryana, India
| | - Anil Kumar Banothu
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Rajendranagar, Hyderabad, 500030, PVNRTVU, Telangana, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, 151401, Punjab, India
| | - Kala Kumar Bharani
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science (CVSc), Warangal, 506166, PVNRTVU, Telangana, India
| |
Collapse
|
11
|
Wu D, Si M, Xue HY, Tran NT, Khalili K, Kaminski R, Wong HL. Lipid nanocarrier targeting activated macrophages for antiretroviral therapy of HIV reservoir. Nanomedicine (Lond) 2023; 18:1343-1360. [PMID: 37815117 PMCID: PMC10652294 DOI: 10.2217/nnm-2023-0120] [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: 04/23/2023] [Accepted: 08/21/2023] [Indexed: 10/11/2023] Open
Abstract
Aim: To develop lipid nano-antiretrovirals (LNAs) for the treatment of HIV-infected macrophages. Materials & methods: LNAs were prepared with docosahexaenoic acid to facilitate brain penetration and surface-decorated with folate considering that infected macrophages often overexpress folate receptors. Results: Folate-decorated LNAs loading rilpivirine (RPV) were efficiently taken up by folate receptor-expressing cell types including activated macrophages. The intracellular Cmax of the RPV-LNAs in activated macrophages was 2.54-fold and the area under the curve was 3.4-fold versus free RPV, translating to comparable or higher (p < 0.01; RPV ≤6.5 ng/ml) activities against HIV infectivity and superior protection (p < 0.05) against HIV cytotoxicity. LNAs were also effective in monocyte-derived macrophages. Conclusion: These findings demonstrate the potential of LNAs for the treatment of infected macrophages, which are key players in HIV reservoirs.
Collapse
Affiliation(s)
- Di Wu
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Mengjie Si
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Hui Yi Xue
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Ngoc T Tran
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Kamel Khalili
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Rafal Kaminski
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ho Lun Wong
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
12
|
Alam A, Ansari MJ, Alqarni MH, Salkini MA, Raish M. Antioxidant, Antibacterial, and Anticancer Activity of Ultrasonic Nanoemulsion of Cinnamomum Cassia L. Essential Oil. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12040834. [PMID: 36840181 PMCID: PMC9966450 DOI: 10.3390/plants12040834] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 06/01/2023]
Abstract
Cinnamomum cassia (C. assia) has long been used in traditional holistic medicine for its medicinal properties. It is used as an antioxidant, antibacterial, anti-inflammatory and anticancer agent. Cinnamon, in particular, the essential oil of C. cassia, has significant biological properties. Despite this, the volatility, stability, and insolubility of C. cassia essential oil (CEO) remain the main disadvantages that limit its application, ultimately affecting its pharmacological efficacy. To find a solution to this problem, we developed the CEO nanoemulsion (CEO-NE). For lipophilic compounds, insoluble nanoemulsion-based formulations are a popular delivery strategy. In this research work, a highly stable dosage form named CEO-NE was successfully developed using polysorbate 80 and water. The findings show that the synthesized CEO has a uniform shape with a PDI of 0.380 and an adequate particle size of 221.8 nm. The antioxidant outcomes show excellent results for CEO-NE compared to CEO against DPPH and hydrogen peroxide. The obtained antibacterial activity of CEO-NE was more efficient than that of CEO against Klebsiella pneumonia (MTCC 8911) with 0.025% and 0.05%, respectively. The CEO-NE preparation was tested against an alveolar lung adenocarcinoma cell line (A549) with an IC50 of 50.21 µg/mL for CEO and 18.05 µg/mL for CEO-NE, respectively. These results are encouraging for future translational studies on CEO-NE use in lung cancer therapy due to its excellent antioxidant, antibacterial, and killing kinetic properties.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammed H. Alqarni
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Ayman Salkini
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
13
|
Asl FD, Mousazadeh M, Taji S, Bahmani A, Khashayar P, Azimzadeh M, Mostafavi E. Nano drug-delivery systems for management of AIDS: liposomes, dendrimers, gold and silver nanoparticles. Nanomedicine (Lond) 2023; 18:279-302. [PMID: 37125616 PMCID: PMC10242436 DOI: 10.2217/nnm-2022-0248] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/08/2023] [Indexed: 05/02/2023] Open
Abstract
AIDS causes increasing mortality every year. With advancements in nanomedicine, different nanomaterials (NMs) have been applied to treat AIDS and overcome its limitations. Among different NMs, nanoparticles (NPs) can act as nanocarriers due to their enhanced solubility, sustained release, targeting abilities and facilitation of drug-dose reductions. This review discusses recent advancements in therapeutics for AIDS/HIV using various NMs, mainly focused on three classifications: polymeric, liposomal and inorganic NMs. Polymeric dendrimers, polyethylenimine-NPs, poly(lactic-co-glycolic acid)-NPs, chitosan and the use of liposomal-based delivery systems and inorganic NPs, including gold and silver NPs, are explored. Recent advances, current challenges and future perspectives on the use of these NMs for better management of HIV/AIDS are also discussed.
Collapse
Affiliation(s)
- Fateme Davarani Asl
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, 88138-33435, Iran
| | - Marziyeh Mousazadeh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-154, Iran
| | - Shirinsadat Taji
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-154, Iran
- Institute for Genetics, University of Cologne, Cologne, D-50674, Germany
| | - Abbas Bahmani
- Institute for Nanoscience & Nanotechnology (INST), Sharif University of Technology, Tehran, 14588-89694, Iran
| | - Patricia Khashayar
- Center for Microsystems Technology, Imec & Ghent University, Ghent, 9050, Belgium
| | - Mostafa Azimzadeh
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, 89195-999, Iran
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
14
|
Zambonino MC, Quizhpe EM, Mouheb L, Rahman A, Agathos SN, Dahoumane SA. Biogenic Selenium Nanoparticles in Biomedical Sciences: Properties, Current Trends, Novel Opportunities and Emerging Challenges in Theranostic Nanomedicine. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:424. [PMID: 36770385 PMCID: PMC9921003 DOI: 10.3390/nano13030424] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
Selenium is an important dietary supplement and an essential trace element incorporated into selenoproteins with growth-modulating properties and cytotoxic mechanisms of action. However, different compounds of selenium usually possess a narrow nutritional or therapeutic window with a low degree of absorption and delicate safety margins, depending on the dose and the chemical form in which they are provided to the organism. Hence, selenium nanoparticles (SeNPs) are emerging as a novel therapeutic and diagnostic platform with decreased toxicity and the capacity to enhance the biological properties of Se-based compounds. Consistent with the exciting possibilities offered by nanotechnology in the diagnosis, treatment, and prevention of diseases, SeNPs are useful tools in current biomedical research with exceptional benefits as potential therapeutics, with enhanced bioavailability, improved targeting, and effectiveness against oxidative stress and inflammation-mediated disorders. In view of the need for developing eco-friendly, inexpensive, simple, and high-throughput biomedical agents that can also ally with theranostic purposes and exhibit negligible side effects, biogenic SeNPs are receiving special attention. The present manuscript aims to be a reference in its kind by providing the readership with a thorough and comprehensive review that emphasizes the current, yet expanding, possibilities offered by biogenic SeNPs in the biomedical field and the promise they hold among selenium-derived products to, eventually, elicit future developments. First, the present review recalls the physiological importance of selenium as an oligo-element and introduces the unique biological, physicochemical, optoelectronic, and catalytic properties of Se nanomaterials. Then, it addresses the significance of nanosizing on pharmacological activity (pharmacokinetics and pharmacodynamics) and cellular interactions of SeNPs. Importantly, it discusses in detail the role of biosynthesized SeNPs as innovative theranostic agents for personalized nanomedicine-based therapies. Finally, this review explores the role of biogenic SeNPs in the ongoing context of the SARS-CoV-2 pandemic and presents key prospects in translational nanomedicine.
Collapse
Affiliation(s)
- Marjorie C. Zambonino
- School of Biological Sciences and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí 100119, Ecuador
| | - Ernesto Mateo Quizhpe
- School of Biological Sciences and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí 100119, Ecuador
| | - Lynda Mouheb
- Laboratoire de Recherche de Chimie Appliquée et de Génie Chimique, Hasnaoua I, Université Mouloud Mammeri, BP 17 RP, Tizi-Ouzou 15000, Algeria
| | - Ashiqur Rahman
- Center for Midstream Management and Science, Lamar University, 211 Redbird Ln., Beaumont, TX 77710, USA
| | - Spiros N. Agathos
- Earth and Life Institute, Catholic University of Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Si Amar Dahoumane
- Department of Chemical Engineering, Polytechnique Montréal, C.P. 6079, Succ. Centre-Ville, Montréal, QC H3C 3A7, Canada
- Department of Chemistry and Biochemistry, Université de Moncton, 18, Ave Antonine-Maillet, Moncton, NB E1A 3E9, Canada
| |
Collapse
|
15
|
Alhalmi A, Amin S, Khan Z, Beg S, Al kamaly O, Saleh A, Kohli K. Nanostructured Lipid Carrier-Based Codelivery of Raloxifene and Naringin: Formulation, Optimization, In Vitro, Ex Vivo, In Vivo Assessment, and Acute Toxicity Studies. Pharmaceutics 2022; 14:1771. [PMID: 36145519 PMCID: PMC9500671 DOI: 10.3390/pharmaceutics14091771] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/12/2022] [Accepted: 08/13/2022] [Indexed: 11/19/2022] Open
Abstract
This work aimed to develop dual drug-loaded nanostructured lipid carriers of raloxifene and naringin (RLX/NRG NLCs) for breast cancer. RLX/NRG NLCs were prepared using Compritol 888 ATO and oleic acid using a hot homogenization-sonication method and optimized using central composite design (CCD). The optimized RLX/NRG NLCs were characterized and evaluated using multiple technological means. The optimized RLX/NRG NLCs exhibited a particle size of 137.12 nm, polydispersity index (PDI) of 0.266, zeta potential (ZP) of 25.9 mV, and entrapment efficiency (EE) of 91.05% (raloxifene) and 85.07% (naringin), respectively. In vitro release (81 ± 2.2% from RLX/NRG NLCs and 31 ± 1.9% from the RLX/NRG suspension for RLX and 93 ± 1.5% from RLX/NRG NLCs and 38 ± 2.01% from the RLX/NRG suspension for NRG within 24 h). Concurrently, an ex vivo permeation study exhibited nearly 2.3 and 2.1-fold improvement in the permeability profiles of RLX and NRG from RLX/NRG NLCs vis-à-vis the RLX/NRG suspension. The depth of permeation was proved with CLSM images which revealed significant permeation of the drug from the RLX/NRG NLCs formulation, 3.5-fold across the intestine, as compared with the RLX/NRG suspension. An in vitro DPPH antioxidant study displayed a better antioxidant potential of RLX/NRG in comparison to RLX and NRG alone due to the synergistic antioxidant effect of RLX and NRG. An acute toxicity study in Wistar rats showed the safety profile of the prepared nanoformulations and their excipients. Our findings shed new light on how poorly soluble and poorly permeable medicines can be codelivered using NLCs in an oral nanoformulation to improve their medicinal performance.
Collapse
Affiliation(s)
- Abdulsalam Alhalmi
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Saima Amin
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Zafar Khan
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sarwar Beg
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Flyde Road, Preston PR1 2HE, UK
| | - Omkulthom Al kamaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
- Lloyd Institute of Management and Technology (Pharm.), Plot No 11, Knowledge Park-II, Greater Noida 201308, India
| |
Collapse
|
16
|
Rojekar S, Abadi LF, Pai R, Prajapati MK, Kulkarni S, Vavia PR. Mannose-Anchored Nano-Selenium Loaded Nanostructured Lipid Carriers of Etravirine for Delivery to HIV Reservoirs. AAPS PharmSciTech 2022; 23:230. [PMID: 35978154 DOI: 10.1208/s12249-022-02377-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022] Open
Abstract
The present investigation aims to develop and explore mannosylated lipid-based carriers to deliver an anti-HIV drug, Etravirine (TMC) and Selenium nanoparticles (SeNPs), to the HIV reservoirs via the mannose receptor. The successful mannosylation was evaluated by the change in zeta potential and lectin binding assay using fluorescence microscopy. Electron microscopy and scattering studies were employed to study the structure and surface of the nanocarrier system. The presence of selenium at the core-shell of the nanocarrier system was confirmed by X-ray photoelectron spectroscopy and energy dispersive X-ray analysis. Further, the in vitro anti-HIV1 efficacy was assessed using HIV1 infected TZM-bl cells followed by in vivo biodistribution studies to evaluate distribution to various reservoirs of HIV. The results exhibited higher effectiveness and a significant increase in the therapeutic index as against the plain drug. The confocal microscopy and flow cytometry studies exhibited the efficient uptake of the coumarin-6 tagged respective formulations. The protective effect of nano selenium toward oxidative stress was evaluated in rats, demonstrating the potential of the lipidic nanoparticle-containing selenium in mitigating oxidative stress in all the major organs. The in vivo biodistribution assessment in rats showed a 12.44, 8.05 and 9.83-fold improvement in the brain, ovary, and lymph node biodistribution, respectively as compared with plain TMC. Delivery of such a combination via mannosylated nanostructured lipid carriers could be an efficient approach for delivering drugs to reservoirs of HIV while simultaneously reducing the oxidative stress induced by such long-term therapies by co-loading Nano-Selenium.
Collapse
Affiliation(s)
- Satish Rojekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400 019, India
| | - Leila Fotooh Abadi
- Department of Virology, Indian Council of Medical Research, National AIDS Research Institute, Pune, 411 026, India
| | - Rohan Pai
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400 056, India
| | - Mahendra Kumar Prajapati
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400 019, India
| | - Smita Kulkarni
- Department of Virology, Indian Council of Medical Research, National AIDS Research Institute, Pune, 411 026, India
| | - Pradeep R Vavia
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, 400 019, India.
| |
Collapse
|
17
|
Fotooh Abadi L, Damiri F, Zehravi M, Joshi R, Pai R, Berrada M, Massoud EES, Rahman MH, Rojekar S, Cavalu S. Novel Nanotechnology-Based Approaches for Targeting HIV Reservoirs. Polymers (Basel) 2022; 14:3090. [PMID: 35956604 PMCID: PMC9370744 DOI: 10.3390/polym14153090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 12/04/2022] Open
Abstract
Highly active anti-retroviral therapy (HAART) is prescribed for HIV infection and, to a certain extent, limits the infection's spread. However, it cannot completely eradicate the latent virus in remote and cellular reservoir areas, and due to the complex nature of the infection, the total eradication of HIV is difficult to achieve. Furthermore, monotherapy and multiple therapies are not of much help. Hence, there is a dire need for novel drug delivery strategies that may improve efficacy, decrease side effects, reduce dosing frequency, and improve patient adherence to therapy. Such a novel strategy could help to target the reservoir sites and eradicate HIV from different biological sanctuaries. In the current review, we have described HIV pathogenesis, the mechanism of HIV replication, and different biological reservoir sites to better understand the underlying mechanisms of HIV spread. Further, the review deliberates on the challenges faced by the current conventional drug delivery systems and introduces some novel drug delivery strategies that have been explored to overcome conventional drug delivery limitations. In addition, the review also summarizes several nanotechnology-based approaches that are being explored to resolve the challenges of HIV treatment by the virtue of delivering a variety of anti-HIV agents, either as combination therapies or by actively targeting HIV reservoir sites.
Collapse
Affiliation(s)
- Leila Fotooh Abadi
- Department of Virology, Indian Council of Medical Research, National AIDS Research Institute, Pune 411026, Maharashtra, India;
| | - Fouad Damiri
- Laboratory of Biomolecules and Organic Synthesis (BIOSYNTHO), Department of Chemistry, Faculty of Sciences Ben M’Sick, University Hassan II of Casablanca, Casablanca 20000, Morocco;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University, Alkharj 11942, Saudi Arabia;
| | - Rohit Joshi
- Precision NanoSystem Inc., Vancouver, BC V6P 6T7, Canada;
| | - Rohan Pai
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India;
| | - Mohammed Berrada
- Laboratory of Biomolecules and Organic Synthesis (BIOSYNTHO), Department of Chemistry, Faculty of Sciences Ben M’Sick, University Hassan II of Casablanca, Casablanca 20000, Morocco;
| | - Ehab El Sayed Massoud
- Biology Department, Faculty of Science and Arts in Dahran Aljnoub, King Khalid University, Abha 62529, Saudi Arabia;
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
- Agriculture Research Centre, Soil, Water and Environment Research Institute, Giza 3725004, Egypt
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Gangwon-do, Wonju 26426, Korea;
| | - Satish Rojekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, Maharashtra, India
- Departments of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
18
|
Nanoparticle-based strategies to target HIV-infected cells. Colloids Surf B Biointerfaces 2022; 213:112405. [PMID: 35255375 DOI: 10.1016/j.colsurfb.2022.112405] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 02/06/2023]
Abstract
Antiretroviral drugs employed for the treatment of human immunodeficiency virus (HIV) infections have remained largely ineffective due to their poor bioavailability, numerous adverse effects, modest uptake in infected cells, undesirable drug-drug interactions, the necessity for long-term drug therapy, and lack of access to tissues and reservoirs. Nanotechnology-based interventions could serve to overcome several of these disadvantages and thereby improve the therapeutic efficacy of antiretrovirals while reducing the morbidity and mortality due to the disease. However, attempts to use nanocarriers for the delivery of anti-retroviral drugs have started gaining momentum only in the past decade. This review explores in-depth the various nanocarriers that have been employed for the treatment of HIV infections highlighting their merits and possible demerits.
Collapse
|