1
|
Ash K, Dev A. Harnessing Nanotechnology in HIV Therapy: Exploring Molecular Pathogenesis and Treatment Strategies with Special Reference to Chemotherapy and Immunotherapy. Microb Pathog 2025; 204:107625. [PMID: 40268149 DOI: 10.1016/j.micpath.2025.107625] [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: 01/02/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
Human immunodeficiency virus (HIV) continues to be a global threat, contributing substantially to social and economic burdens worldwide. Synthetic ARV drugs are classified into six different classes viz NRTIs, NNRTIs, PIs, IIs, INSTIs, and FIs. Highly active anti-retroviral therapy (HAART) which is a combination of two or more ARV drugs from different classes is gaining immense popularity in the HIV therapy regimen due to its better therapeutic outcome. However, despite its successful endeavor in significant viral suppression, synthetic drugs are associated with numerous adverse effects. To mitigate this issue, scientists are exploring ARV agents derived from various natural sources like plants, and marine organisms that can exhibit potent anti-HIV activity with minimal side effects. Nevertheless, both synthetically and naturally derived ARV agents have failed to exhibit eradication of HIV from latent reservoirs. Henceforth, researchers are shifting their attention towards formulating a drug-encapsulated nano-delivery system to ensure a significant amount of drug delivery into these reservoirs. Additionally, the discovery of a novel HIV vaccine that can induce robust immune responses against multiple HIV strains and facilitate complete removal of the virus before the establishment of a latent reservoir is the need of an hour. Briefly, we discussed various synthetic and natural chemotherapeutic agents along with their specificity and limitations, different drug-delivery devices for ART, immunotherapy, vaccines, and lastly, challenges and strategies associated with vaccine development.
Collapse
Affiliation(s)
- Kaushiki Ash
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| |
Collapse
|
2
|
Kour P, Saha P, Bhattacharya S, Kumari D, Debnath A, Roy A, Sharma DK, Mukherjee D, Singh K. Design, synthesis, and biological evaluation of 3,3'-diindolylmethane N-linked glycoconjugate as a leishmanial topoisomerase IB inhibitor with reduced cytotoxicity. RSC Med Chem 2023; 14:2100-2114. [PMID: 37859718 PMCID: PMC10583832 DOI: 10.1039/d3md00214d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/25/2023] [Indexed: 10/21/2023] Open
Abstract
Leishmaniasis, one of the neglected diseases, ranks second to malaria in the cause of parasitic mortality and morbidity. The present chemotherapeutic regimen faces the limitations of drug resistance and toxicity concerns, raising a great need to develop new chemotherapeutic leads that are orally administrable, potent, non-toxic, and cost-effective. Several research groups came forward to fill this therapeutic gap with new classes of active compounds against leishmaniasis, one such being 3,3'-diindolylmethane (DIM) derivatives. We tried to link this concept with another promising approach of glycoconjugation to study how glycosylated groups work differently from non-glycosylated ones. In the present study, a series of 3,3'-DIM derivatives have been synthesized and screened for their anti-leishmanial potency on Leishmania donovani promastigotes. Next, we synthesized the β-N,N' glycoside of potent compound 3d using indole-indoline conversion, Fischer-type glycosylation, 2,3-dichloro-5,6-dicyano-1,4-benzoquionone (DDQ) oxidation, and molecular iodine catalyzed coupling with a suitable aldehyde in reasonable overall yield. The biological evaluation revealed that glycosides had reduced cytotoxic effects on the J774A.1 macrophage cell line. The enzyme inhibition study confirms that the glycoside derivatives have significant inhibitory activity against the leishmanial topoisomerase IB enzyme. Molecular docking further displayed the better binding efficiency of glycoside 13 with the target enzyme, suggesting the involvement of more H-bond interactions in the case of glycosides as compared to free drugs. Therefore, this work helps in proposing the fact that the addition of sugar moieties adds some favorable characteristics to free inhibitors, making it a promising approach for future clinical diagnostic and therapeutic applications, which can prove to be a valuable arsenal in combating such neglected diseases.
Collapse
Affiliation(s)
- Parampreet Kour
- Infectious Diseases Division, CSIR - Indian Institute of Integrative Medicine Canal Road Jammu 180001 India +91 191 2585006 13 Ext: 333
| | - Pallavi Saha
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology Banaras Hindu University Varanasi-221005 India
| | - Srija Bhattacharya
- Natural Products & Medicinal Chemistry Division, CSIR - Indian Institute of Integrative Medicine Jammu 180001 India
| | - Diksha Kumari
- Infectious Diseases Division, CSIR - Indian Institute of Integrative Medicine Canal Road Jammu 180001 India +91 191 2585006 13 Ext: 333
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| | - Abhipsa Debnath
- Department of Biotechnology, Savitribai Phule Pune University Pune-411007 India
| | - Amit Roy
- Department of Biotechnology, Savitribai Phule Pune University Pune-411007 India
| | - Deepak K Sharma
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology Banaras Hindu University Varanasi-221005 India
| | - Debaraj Mukherjee
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
- Department of Chemical Sciences, Unified Academic Campus, Bose Institute Kolkata 700091 India
| | - Kuljit Singh
- Infectious Diseases Division, CSIR - Indian Institute of Integrative Medicine Canal Road Jammu 180001 India +91 191 2585006 13 Ext: 333
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad-201002 India
| |
Collapse
|
3
|
Rocamora F, Peralta AG, Shin S, Sorrentino J, Wu MYM, Toth EA, Fuerst TR, Lewis NE. Glycosylation shapes the efficacy and safety of diverse protein, gene and cell therapies. Biotechnol Adv 2023; 67:108206. [PMID: 37354999 PMCID: PMC11168894 DOI: 10.1016/j.biotechadv.2023.108206] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Over recent decades, therapeutic proteins have had widespread success in treating a myriad of diseases. Glycosylation, a near universal feature of this class of drugs, is a critical quality attribute that significantly influences the physical properties, safety profile and biological activity of therapeutic proteins. Optimizing protein glycosylation, therefore, offers an important avenue to developing more efficacious therapies. In this review, we discuss specific examples of how variations in glycan structure and glycoengineering impacts the stability, safety, and clinical efficacy of protein-based drugs that are already in the market as well as those that are still in preclinical development. We also highlight the impact of glycosylation on next generation biologics such as T cell-based cancer therapy and gene therapy.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Angelo G Peralta
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seunghyeon Shin
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - James Sorrentino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mina Ying Min Wu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
4
|
Žigrayová D, Mikušová V, Mikuš P. Advances in Antiviral Delivery Systems and Chitosan-Based Polymeric and Nanoparticulate Antivirals and Antiviral Carriers. Viruses 2023; 15:647. [PMID: 36992356 PMCID: PMC10054433 DOI: 10.3390/v15030647] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Current antiviral therapy research is focused on developing dosage forms that enable highly effective drug delivery, providing a selective effect in the organism, lower risk of adverse effects, a lower dose of active pharmaceutical ingredients, and minimal toxicity. In this article, antiviral drugs and the mechanisms of their action are summarized at the beginning as a prerequisite background to develop relevant drug delivery/carrier systems for them, classified and briefly discussed subsequently. Many of the recent studies aim at different types of synthetic, semisynthetic, and natural polymers serving as a favorable matrix for the antiviral drug carrier. Besides a wider view of different antiviral delivery systems, this review focuses on advances in antiviral drug delivery systems based on chitosan (CS) and derivatized CS carriers. CS and its derivatives are evaluated concerning methods of their preparation, their basic characteristics and properties, approaches to the incorporation of an antiviral drug in the CS polymer as well as CS nanoparticulate systems, and their recent biomedical applications in the context of actual antiviral therapy. The degree of development (i.e., research study, in vitro/ex vivo/in vivo preclinical testing), as well as benefits and limitations of CS polymer and CS nanoparticulate drug delivery systems, are reported for particular viral diseases and corresponding antivirotics.
Collapse
Affiliation(s)
- Dominika Žigrayová
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
| | - Veronika Mikušová
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
| | - Peter Mikuš
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
| |
Collapse
|
5
|
Aanish Ali M, Rehman N, Park TJ, Basit MA. Antiviral role of nanomaterials: a material scientist's perspective. RSC Adv 2022; 13:47-79. [PMID: 36605642 PMCID: PMC9769549 DOI: 10.1039/d2ra06410c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
The present world continues to face unprecedented challenges caused by the COVID-19 pandemic. Collaboration between researchers of multiple disciplines is the need of the hour. There is a need to develop antiviral agents capable of inhibiting viruses and tailoring existing antiviral drugs for efficient delivery to prevent a surge in deaths caused by viruses globally. Biocompatible systems have been designed using nanotechnological principles which showed appreciable results against a wide range of viruses. Many nanoparticles can act as antiviral therapeutic agents if synthesized by the correct approach. Moreover, nanoparticles can act as carriers of antiviral drugs while overcoming their inherent drawbacks such as low solubility, poor bioavailability, uncontrolled release, and side effects. This review highlights the potential of nanomaterials in antiviral applications by discussing various studies and their results regarding antiviral potential of nanoparticles while also suggesting future directions to researchers.
Collapse
Affiliation(s)
- Muhammad Aanish Ali
- Department of Materials Science and Engineering, Institute of Space Technology Islamabad 44000 Pakistan
| | - Nagina Rehman
- Department of Zoology, Government College University Allama Iqbal Road Faisalabad 38000 Pakistan
| | - Tae Joo Park
- Department of Materials Science and Chemical Engineering, Hanyang University Ansan 15588 Republic of Korea
| | - Muhammad Abdul Basit
- Department of Materials Science and Engineering, Institute of Space Technology Islamabad 44000 Pakistan
| |
Collapse
|
6
|
Zlotnikov ID, Kudryashova EV. Mannose Receptors of Alveolar Macrophages as a Target for the Addressed Delivery of Medicines to the Lungs. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022010150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
7
|
Lipid Nanocarriers for Anti-HIV Therapeutics: A Focus on Physicochemical Properties and Biotechnological Advances. Pharmaceutics 2021; 13:pharmaceutics13081294. [PMID: 34452255 PMCID: PMC8398060 DOI: 10.3390/pharmaceutics13081294] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/03/2021] [Accepted: 08/07/2021] [Indexed: 12/13/2022] Open
Abstract
Since HIV was first identified, and in a relatively short period of time, AIDS has become one of the most devastating infectious diseases of the 21st century. Classical antiretroviral therapies were a major step forward in disease treatment options, significantly improving the survival rates of HIV-infected individuals. Even though these therapies have greatly improved HIV clinical outcomes, antiretrovirals (ARV) feature biopharmaceutic and pharmacokinetic problems such as poor aqueous solubility, short half-life, and poor penetration into HIV reservoir sites, which contribute to the suboptimal efficacy of these regimens. To overcome some of these issues, novel nanotechnology-based strategies for ARV delivery towards HIV viral reservoirs have been proposed. The current review is focused on the benefits of using lipid-based nanocarriers for tuning the physicochemical properties of ARV to overcome biological barriers upon administration. Furthermore, a correlation between these properties and the potential therapeutic outcomes has been established. Biotechnological advancements using lipid nanocarriers for RNA interference (RNAi) delivery for the treatment of HIV infections were also discussed.
Collapse
|
8
|
Martínez-Segoviano IDJ, Ganem-Rondero A. Enhancement of the transdermal delivery of zidovudine by pretreating the skin with two physical enhancers: microneedles and sonophoresis. ACTA ACUST UNITED AC 2021; 29:279-290. [PMID: 34216369 DOI: 10.1007/s40199-021-00402-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/25/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Zidovudine (AZT) has been the most widely used drug for antiretroviral therapy. In order to improve the therapy with this drug, different alternatives have been proposed, such as the transdermal administration. The use of permeation enhancers is necessary to favor the passage of this drug through the skin, due to its physicochemical properties and to the natural permeation barrier imposed by the skin. OBJECTIVES To evaluate the effect of two permeation enhancers, sonophoresis and microneedles, on the permeability of AZT through the skin. METHODS Permeation studies with an AZT solution were performed using pigskin clamped in Franz-type cells. Sonophoresis was applied under different conditions (i.e., amplitude, duty cycle and application time), selected according to an experimental design, where the response variables were the increase in temperature of the skin surface and the increase in transepidermal water loss. ATR-FTIR was also used to demonstrate the effect of enhancers on membrane components. RESULTS The permeability of AZT through intact skin was very poor, with a very long lag time. Pretreatment of the skin with sonophoresis increased AZT transport significantly, reducing the lag time. The maximum flux (27.52 µgcm-2 h-1) and the highest total amount permeated (about 624 µg/cm2) were obtained when applying sonophoresis in continuous mode, with an amplitude of 20%, and an application time of 2 min. Sonophoresis appears to have an impact on stratum corneum proteins. The use of microneedles further increased the flux (30.41 µgcm-2 h-1) and the total amount permeated (about 916 µg/cm2), relative to sonophoresis. CONCLUSION The results are encouraging in terms of promoting AZT transport through the skin using sonophoresis or microneedles as permeation enhancers.
Collapse
Affiliation(s)
- Irene de Jesús Martínez-Segoviano
- División de Estudios de Posgrado (Tecnología Farmacéutica), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Av. 1o de Mayo s/n, 54740, Cuautitlán Izcalli, Estado de México, Mexico
| | - Adriana Ganem-Rondero
- División de Estudios de Posgrado (Tecnología Farmacéutica), Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Av. 1o de Mayo s/n, 54740, Cuautitlán Izcalli, Estado de México, Mexico.
| |
Collapse
|
9
|
Al-Sawaftah NM, Abusamra RH, Husseini GA. Carbohydrate-functionalized Liposomes in Cancer Therapy. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716999200626144921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Existing cancer treatments are often accompanied by adverse side effects that can greatly
reduce the quality of life of cancer patients; this sets the platform for the development and application
of nanocarrier-based platforms for the delivery of anticancer drugs. Among these nanocarriers,
liposomes have demonstrated excellent potential in drug delivery applications. Furthermore,
the overexpression of certain receptors on cancer cells has led to the development of active targeting
approaches where liposome surfaces are decorated with ligands against these receptors. Given
the central role that sugars play in cancer biology, more and more researchers are integrating “glycoscience”
into their anticancer therapeutic designs. Carbohydrate functionalized liposomes present
an attractive drug delivery system due to their biocompatibility, biodegradability, low toxicity,
and specific cell targeting ability. This review presents an overview of the preparation methods,
characterization, evaluation, and applications of carbohydrate functionalized liposomes in cancer
therapy.
Collapse
Affiliation(s)
- Nour M. Al-Sawaftah
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Rand H. Abusamra
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
10
|
Stevens DM, Adiseshaiah P, Dasa SSK, Potter TM, Skoczen SL, Snapp KS, Cedrone E, Patel N, Busman-Sahay K, Rosen EP, Sykes C, Cottrell M, Dobrovolskaia MA, Estes JD, Kashuba ADM, Stern ST. Application of a Scavenger Receptor A1-Targeted Polymeric Prodrug Platform for Lymphatic Drug Delivery in HIV. Mol Pharm 2020; 17:3794-3812. [PMID: 32841040 PMCID: PMC7861197 DOI: 10.1021/acs.molpharmaceut.0c00562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We have developed a macromolecular prodrug platform based on poly(l-lysine succinylated) (PLS) that targets scavenger receptor A1 (SR-A1), a receptor expressed by myeloid and endothelial cells. We demonstrate the selective uptake of PLS by murine macrophage, RAW 264.7 cells, which was eliminated upon cotreatment with the SR-A inhibitor polyinosinic acid (poly I). Further, we observed no uptake of PLS in an SR-A1-deficient RAW 264.7 cell line, even after 24 h incubation. In mice, PLS distributed to lymphatic organs following i.v. injection, as observed by ex vivo fluorescent imaging, and accumulated in lymph nodes following both i.v. and i.d. administrations, based on immunohistochemical analysis with high-resolution microscopy. As a proof-of-concept, the HIV antiviral emtricitabine (FTC) was conjugated to the polymer's succinyl groups via ester bonds, with a drug loading of 14.2% (wt/wt). The prodrug (PLS-FTC) demonstrated controlled release properties in vitro with a release half-life of 15 h in human plasma and 29 h in esterase-inhibited plasma, indicating that drug release occurs through both enzymatic and nonenzymatic mechanisms. Upon incubation of PLS-FTC with human peripheral blood mononuclear cells (PBMCs), the released drug was converted to the active metabolite FTC triphosphate. In a pharmacokinetic study in rats, the prodrug achieved ∼7-19-fold higher concentrations in lymphatic tissues compared to those in FTC control, supporting lymphatic-targeted drug delivery. We believe that the SR-A1-targeted macromolecular PLS prodrug platform has extraordinary potential for the treatment of infectious diseases.
Collapse
Affiliation(s)
- David M Stevens
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Pavan Adiseshaiah
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Siva S K Dasa
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Tim M Potter
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Sarah L Skoczen
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Kelsie S Snapp
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Edward Cedrone
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Nimit Patel
- Small Animal Imaging Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
| | - Elias P Rosen
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Craig Sykes
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mackenzie Cottrell
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| | - Jacob D Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon 97006, United States
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon 97239, United States
| | - Angela D M Kashuba
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Stephan T Stern
- Nanotechnology Characterization Lab, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, Maryland 21702-1201, United States
| |
Collapse
|
11
|
Khan T, Mayuresh Patkar M, Momin M, Omri A. Macrophage targeted nanocarrier delivery systems in HIV therapeutics. Expert Opin Drug Deliv 2020; 17:903-918. [PMID: 32347124 DOI: 10.1080/17425247.2020.1762565] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Human immunodeficiency virus (HIV) targets and modulates the immune system increasing the risk of other associated infections. Highly active antiretroviral therapy (HAART) has significantly improved AIDS-associated morbidity, but has limitations of adverse effects, frequent dosing regimen leading to medical non-adherence. Drug delivery systems that target HIV reservoirs could potentially reduce dose-dependent toxicity and the duration of treatment. The major cellular HIV reservoirs are macrophages and CD4+ T cells with macrophages being responsible for carrying and spreading the virus. The crucial involvement of macrophages in the pathogenesis of HIV infection has led to development of macrophage targeted nanocarrier delivery systems. AREAS COVERED Eradication of viral reservoirs like HIV-infected macrophages has emerged to be a fundamental barrier and challenge for complete eradication of HIV from the immune system. Literature reports several macrophage targeted nanocarrier delivery systems developed as either functionalized or non-functionalized formulations such as liposomes, ethosomes, polymeric nanoparticles, dendrimers, and solid lipid nanoparticles showcasing superior efficacy over the conventional antiretroviral delivery systems. EXPERT OPINION The development of fixed dose combination of antiretroviral drugs into macrophage targeted delivery systems should factor in the inherent plasticity and heterogeneity of macrophages that is dependent on their microenvironment. A rational selection of nanocarriers will facilitate selectivity and enhanced efficacy of antiretroviral drugs accompanied by reduced dosing and toxicity. Such macrophage targeted delivery systems would positively impact the therapeutic outcomes in the management of HIV infection.
Collapse
Affiliation(s)
- Tabassum Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Mayuresh Mayuresh Patkar
- Department of Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy , Mumbai, Maharashtra, India
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University , Sudbury, ON, Canada
| |
Collapse
|
12
|
Arruda EL, Japiassu KB, de Melo Souza PL, Araújo KCF, Thomaz DV, Cortez AP, Garcia LF, Valadares MC, de Souza Gil E, de Oliveira V. Zidovudine Glycosylation by Filamentous Fungi Leads to a Better Redox Stability and Improved Cytotoxicity in B16F10 Murine Melanoma Cells. Anticancer Agents Med Chem 2020; 20:1688-1694. [PMID: 32329702 DOI: 10.2174/1871520620666200424112504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 03/15/2020] [Accepted: 03/15/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The strategic development of therapeutic agents, capable of being targeted at their active sites, has been a major goal in treatment of cancer. The delivery of drugs for tumors has as its main challenge the development of safe and effective drugs, since the goal of chemotherapy is to eliminate the tumor completely without affecting healthy cells. The aim of present study was to investigate the antioxidant, anticancer activities of zidovudine and its α-O-glycosylated derivative obtained by biosynthesis of a filamentous fungi, Cunninghamela echinulata. METHODS An evaluation of the cytotoxic potential of zidovudine and its α-O-glycosylated was performed in fibroblasts and melanoma cells by the tetrazolium reduction method (MTT) and the antioxidant activity of this derivative was observed. RESULTS The antioxidant activity of zidovudine demonstrated an electrochemical oxidation potential of 0.91V, while the α-O-glycosylated derivative did not exhibit any antioxidant activity. The zidovudine exhibited low cytotoxicity for melanoma and fibroblast cells, while the α-O-glycosylated derivative presented better cytotoxicity on melanoma cells at a concentration of 10mg. mL-1. CONCLUSION This study demonstrates the specific cytotoxicity of the glycoconjugate and suggests that glycosylation by biosynthesis can be a useful strategy for obtaining new anticancer compounds.
Collapse
Affiliation(s)
- Evilanna L Arruda
- Laboratorio de Bioconversao, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Kamila B Japiassu
- Laboratorio de Bioconversao, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Paula L de Melo Souza
- Laboratorio de Bioconversao, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Kelly C F Araújo
- Laboratorio de Bioconversao, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Douglas V Thomaz
- Laboratorio de Pesquisa em Produtos Naturais, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, , Goiania, GO, Brazil
| | - Alane P Cortez
- Laboratorio de Farmacologia e Toxicologia Celular, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Luane F Garcia
- Laboratorio de Anlises Farmaceuticas e Ambientais, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Marize C Valadares
- Laboratorio de Farmacologia e Toxicologia Celular, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Eric de Souza Gil
- Laboratorio de Anlises Farmaceuticas e Ambientais, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| | - Valéria de Oliveira
- Laboratorio de Bioconversao, Faculdade de Farmacia, Universidade Federal de Goias, P.O. Box 131, Goiania, GO, Brazil
| |
Collapse
|
13
|
Yavuz B, Morgan JL, Showalter L, Horng KR, Dandekar S, Herrera C, LiWang P, Kaplan DL. Pharmaceutical Approaches to HIV Treatment and Prevention. ADVANCED THERAPEUTICS 2018; 1:1800054. [PMID: 32775613 PMCID: PMC7413291 DOI: 10.1002/adtp.201800054] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Indexed: 12/17/2022]
Abstract
Human immunodeficiency virus (HIV) infection continues to pose a major infectious disease threat worldwide. It is characterized by the depletion of CD4+ T cells, persistent immune activation, and increased susceptibility to secondary infections. Advances in the development of antiretroviral drugs and combination antiretroviral therapy have resulted in a remarkable reduction in HIV-associated morbidity and mortality. Antiretroviral therapy (ART) leads to effective suppression of HIV replication with partial recovery of host immune system and has successfully transformed HIV infection from a fatal disease to a chronic condition. Additionally, antiretroviral drugs have shown promise for prevention in HIV pre-exposure prophylaxis and treatment as prevention. However, ART is unable to cure HIV. Other limitations include drug-drug interactions, drug resistance, cytotoxic side effects, cost, and adherence. Alternative treatment options are being investigated to overcome these challenges including discovery of new molecules with increased anti-viral activity and development of easily administrable drug formulations. In light of the difficulties associated with current HIV treatment measures, and in the continuing absence of a cure, the prevention of new infections has also arisen as a prominent goal among efforts to curtail the worldwide HIV pandemic. In this review, the authors summarize currently available anti-HIV drugs and their combinations for treatment, new molecules under clinical development and prevention methods, and discuss drug delivery formats as well as associated challenges and alternative approaches for the future.
Collapse
Affiliation(s)
- Burcin Yavuz
- Department of Biomedical Engineering Tufts University 4 Colby Street, Medford, MA 02155, USA
| | - Jessica L Morgan
- Department of Molecular Cell Biology University of California-Merced5200 North Lake Road, Merced, CA 95343, USA
| | - Laura Showalter
- Department of Molecular Cell Biology University of California-Merced5200 North Lake Road, Merced, CA 95343, USA
| | - Katti R Horng
- Department of Medical Microbiology and Immunology University of California-Davis 5605 GBSF, 1 Shields Avenue, Davis, CA 95616, USA
| | - Satya Dandekar
- Department of Medical Microbiology and Immunology University of California-Davis 5605 GBSF, 1 Shields Avenue, Davis, CA 95616, USA
| | - Carolina Herrera
- Department of Medicine St. Mary's Campus Imperial College Room 460 Norfolk Place, London W2 1PG, UK
| | - Patricia LiWang
- Department of Molecular Cell Biology University of California-Merced5200 North Lake Road, Merced, CA 95343, USA
| | - David L Kaplan
- Department of Biomedical Engineering Tufts University 4 Colby Street, Medford, MA 02155, USA
| |
Collapse
|
14
|
Monroe M, Flexner C, Cui H. Harnessing nanostructured systems for improved treatment and prevention of HIV disease. Bioeng Transl Med 2018; 3:102-123. [PMID: 30065966 PMCID: PMC6063869 DOI: 10.1002/btm2.10096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/12/2022] Open
Abstract
Combination antiretroviral therapy effectively controls human immunodeficiency virus (HIV) viral replication, delaying the progression to acquired immune deficiency syndrome and improving and extending quality of life of patients. However, the inability of antiretroviral therapeutics to target latent virus and their poor penetration of viral reserve tissues result in the need for continued treatment for the life of the patient. Side effects from long-term antiretroviral use and the development of drug resistance due to patient noncompliance are also continuing problems. Nanostructured systems of antiretroviral therapeutics have the potential to improve targeted delivery to viral reservoirs, reduce drug toxicity, and increase dosing intervals, thereby improving treatment outcomes and enhancing patient adherence. Despite these advantages, very few nanostructured antiretroviral delivery systems have made it to clinical trials due to challenges in preclinical and clinical development. In this context, we review the current challenges in HIV disease management, and the recent progress in leveraging the unique performance of nanostructured systems in therapeutic delivery for improved treatment and prevention of this incurable human disease.
Collapse
Affiliation(s)
- Maya Monroe
- Dept. of Chemical and Biomolecular Engineering The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218.,Institute for NanoBioTechnology The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218
| | - Charles Flexner
- Div. of Clinical Pharmacology and Infectious Diseases Johns Hopkins University School of Medicine and Bloomberg School of Public Health Baltimore MD 21205
| | - Honggang Cui
- Dept. of Chemical and Biomolecular Engineering The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218.,Institute for NanoBioTechnology The Johns Hopkins University, 3400 N Charles Street Baltimore MD 21218.,Dept. of Oncology, Sidney Kimmel Comprehensive Cancer Center The Johns Hopkins University School of Medicine Baltimore MD 21205.,Center for Nanomedicine The Wilmer Eye Institute, The Johns Hopkins University School of Medicine Baltimore MD 21231
| |
Collapse
|
15
|
Cai L, Gu Z, Zhong J, Wen D, Chen G, He L, Wu J, Gu Z. Advances in glycosylation-mediated cancer-targeted drug delivery. Drug Discov Today 2018; 23:1126-1138. [DOI: 10.1016/j.drudis.2018.02.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/22/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022]
|
16
|
Abstract
The phenomenal advances in pharmaceutical sciences over the last few decades have led to the development of new therapeutics like peptides, proteins, RNAs, DNAs and highly potent small molecules. Fruitful applications of these therapeutics have been challenged by several anatomical and physiological barriers that limit adequate drug disposition at the site-of-action and by off-target drug distribution to undesired tissues, which together result in the reduced effectiveness and increased side effects of therapeutic agents. As such, the development of drug delivery and targeting systems has been recognised as a cornerstone for future drug development. Research in pharmaceutical sciences is now devoted to tackling delivery challenges through engineering delivery systems that move beyond conventional dosage forms and regimens into state-of-the-art targeted drug delivery tailored toward specific therapeutic needs. Modern drug delivery systems comprise passive and active targeting approaches. While passive targeting relies on the natural course of distribution of drugs or drug carriers in the body, as governed by their physicochemical properties, active targeting often exploits targeting moieties that home preferentially into target tissues. Here, we provide an overview of theories of and approaches to passive and active drug delivery. As the design of drug delivery is dependent on the unique structure of target tissues and organs, we present our discussion in an organ-specific manner with the aim to inspire the development of new strategies for curing disease with high accuracy and efficiency.
Collapse
Affiliation(s)
- Mohammad Alsaggar
- a Department of Pharmaceutical Technology, College of Pharmacy , Jordon University of Science and Technology , Irbid , Jordan
| | - Dexi Liu
- b Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy , University of Georgia , Athens , GA , USA
| |
Collapse
|
17
|
Lembo D, Donalisio M, Civra A, Argenziano M, Cavalli R. Nanomedicine formulations for the delivery of antiviral drugs: a promising solution for the treatment of viral infections. Expert Opin Drug Deliv 2017; 15:93-114. [DOI: 10.1080/17425247.2017.1360863] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- David Lembo
- Department of Clinical and Biological Sciences, University of Torino, S. Luigi Gonzaga Hospital, Torino, Italy
| | - Manuela Donalisio
- Department of Clinical and Biological Sciences, University of Torino, S. Luigi Gonzaga Hospital, Torino, Italy
| | - Andrea Civra
- Department of Clinical and Biological Sciences, University of Torino, S. Luigi Gonzaga Hospital, Torino, Italy
| | - Monica Argenziano
- Department of Drug Science and Technology, University of Torino, Turin, Italy
| | - Roberta Cavalli
- Department of Drug Science and Technology, University of Torino, Turin, Italy
| |
Collapse
|
18
|
Augmented delivery of gemcitabine in lung cancer cells exploring mannose anchored solid lipid nanoparticles. J Colloid Interface Sci 2016; 481:107-16. [DOI: 10.1016/j.jcis.2016.07.020] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/10/2016] [Accepted: 07/11/2016] [Indexed: 12/11/2022]
|
19
|
Park HJ, Jeon EJ, Lee JS, Hong SH, Cho AN, Lee J, Moon JS, Jung KE, Oh JW, Lee H, Cho SW. Galactosylated Lipidoid Nanoparticles for Delivery of Small Interfering RNA to Inhibit Hepatitis C Viral Replication In Vivo. Adv Healthc Mater 2016; 5:2931-2941. [PMID: 27774775 DOI: 10.1002/adhm.201600416] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/17/2016] [Indexed: 01/08/2023]
Abstract
Small interfering RNA (siRNA) delivery can provide an effective therapy for treating viral diseases by silencing genes involved in viral replication. In this study, a liver-targeting formulation of lipidoid nanoparticle for delivery of siRNA that targets protein kinase C-related kinase 2 (PRK2) to inhibit hepatitis C virus (HCV) replication is reported. The most effective, minimally cytotoxic lipidoid for siRNA delivery to hepatic cells is identified from a small library of alkyl epoxide-polyamine conjugates. In vitro transfection of PRK2 siRNA (siPRK2) using this lipidoid induces significant silencing of PRK2 (≈80%), suppressing HCV replication in human hepatic cells transfected with the HCV subgenomic replicon. Systemic administration of siPRK2 using the lipidoid nanoparticles results in significant reduction of host PRK2 in the mouse liver (≈60%). This treatment significantly suppresses HCV replication in an HCV-xenograft mouse model. siRNA delivery to the liver is further improved via galactosylation of the lipidoid. Compared with the unmodified lipidoid formulation, galactosylated lipidoids induce greater silencing of host PRK2 in mouse livers (≈80%) and more rapid suppression of HCV replication in an HCV-xenograft mouse. This study suggests that galactosylated lipidoid nanoparticles could provide a treatment for hepatitis C by mediating delivery of anti-viral RNA interference therapeutics to the liver.
Collapse
Affiliation(s)
- Hyun-Ji Park
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| | - Eun Je Jeon
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| | - Jung Seung Lee
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| | - Sang Hyeon Hong
- Department of Chemistry; The Graduate School of Nanoscience and Technology; Korea Advanced Institute of Science and Technology; 291 Daehak-ro Yuseong-gu, Daejeon 305-701 South Korea
| | - Ann-Na Cho
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| | - Joan Lee
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| | - Jae-Su Moon
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| | - Kyeong-Eun Jung
- Research Center; ST Pharm Co. Ltd; 231 Hyeomnyeok-ro Siheung 135-735 South Korea
| | - Jong-Won Oh
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| | - Haeshin Lee
- Department of Chemistry; The Graduate School of Nanoscience and Technology; Korea Advanced Institute of Science and Technology; 291 Daehak-ro Yuseong-gu, Daejeon 305-701 South Korea
| | - Seung-Woo Cho
- Department of Biotechnology; Yonsei University; 50 Yonsei-ro Seodaemun-guSeoul 120-749 South Korea
| |
Collapse
|
20
|
Preparation of complementary glycosylated hyperbranched polymer/poly(ethylene glycol) brushes and their selective interactions with hepatocytes. Colloids Surf B Biointerfaces 2016; 145:309-318. [DOI: 10.1016/j.colsurfb.2016.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/13/2016] [Accepted: 05/04/2016] [Indexed: 01/05/2023]
|
21
|
L. Arruda E, S. Nunes E, de M. Souza PL, B. Japiassú K, C. de Carvalho T, G. Vaz B, M. Lião L, de Oliveira V. A single-step O-glycosylation of azidothymidine in bioreactor catalysed by filamentous fungi. Tetrahedron Lett 2016. [DOI: 10.1016/j.tetlet.2016.08.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
22
|
Siram K, Marslin G, Raghavan CV, Balakumar K, Rahman H, Franklin G. A brief perspective on the diverging theories of lymphatic targeting with colloids. Int J Nanomedicine 2016; 11:2867-72. [PMID: 27366065 PMCID: PMC4913961 DOI: 10.2147/ijn.s105852] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
For targeted delivery of colloids to the lymphatic system, the colloids should efficiently reach and remain in the lymphatics for a considerable period of time. As per the current knowledge, diffusion and phagocytosis are the two mechanisms through which colloids reach the lymphatic system. Several parameters including particle size and charge have been shown to affect the direct uptake of colloids by the lymphatic system. Although many researchers attached ligands on the surface of colloids to promote phagocytosis-mediated lymphatic delivery, another school of thought suggests avoidance of phagocytosis by use of carriers like polyethylene glycol (PEG)ylated colloids to impart stealth attributes and evade phagocytosis. In this perspective, we weigh up the paradoxical theories and approaches available in the literature to draw conclusions on the conditions favorable for achieving efficient lymphatic targeting of colloids.
Collapse
Affiliation(s)
- Karthik Siram
- Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore, India
| | - Gregory Marslin
- Centre for the Research and Technology of Agro-Environment and Biological Sciences, University of Minho, Braga, Portugal
| | | | | | - Habibur Rahman
- Department of Pharmaceutics, PSG College of Pharmacy, Coimbatore, India
| | - Gregory Franklin
- Department of Integrative Plant Biology, Institute of Plant Genetics, Polish Academy of Sciences, Poznań, Poland
| |
Collapse
|
23
|
Mauceri A, Fracassi A, D'Abramo M, Borocci S, Giansanti L, Piozzi A, Galantini L, Martino A, D'Aiuto V, Mancini G. Role of the hydrophilic spacer of glucosylated amphiphiles included in liposome formulations in the recognition of Concanavalin A. Colloids Surf B Biointerfaces 2015; 136:232-9. [DOI: 10.1016/j.colsurfb.2015.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 10/23/2022]
|
24
|
Kumar P, Lakshmi YS, C. B, Golla K, Kondapi AK. Improved Safety, Bioavailability and Pharmacokinetics of Zidovudine through Lactoferrin Nanoparticles during Oral Administration in Rats. PLoS One 2015; 10:e0140399. [PMID: 26461917 PMCID: PMC4604150 DOI: 10.1371/journal.pone.0140399] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 09/24/2015] [Indexed: 11/19/2022] Open
Abstract
Zidovudine (AZT) is one of the most referred antiretroviral drug. In spite of its higher bioavailability (50–75%) the most important reason of its cessation are bone marrow suppression, anemia, neutropenia and various organs related toxicities. This study aims at the improvement of oral delivery of AZT through its encapsulation in lactoferrin nanoparticles (AZT-lactonano). The nanoparticles (NPs) are of 50–60 nm in size and exhibit 67% encapsulation of the AZT. They are stable in simulated gastric and intestinal fluids. Anti-HIV-1 activity of AZT remains unaltered in nanoformulation in acute infection. The bioavailability and tissue distribution of AZT is higher in blood followed by liver and kidney. AZT-lactonano causes the improvement of pharmacokinetic profile as compared to soluble AZT; a more than 4 fold increase in AUC and AUMC in male and female rats. The serum Cmax for AZT-lactonano was increased by 30%. Similarly there was nearly 2-fold increase in Tmax and t1/2. Our in vitro study confirms that, the endosomal pH is ideal for drug release from NPs and shows constant release from up to 96h. Bone marrow micronucleus assay show that nanoformulation exhibits approximately 2fold lower toxicity than soluble form. Histopathological and biochemical analysis further confirms that less or no significant organ toxicities when nanoparticles were used. AZT-lactonano has shown its higher efficacy, low organs related toxicities, improved pharmacokinetics parameter while keeping the antiviral activity intact. Thus, the nanoformulation are safe for the target specific drug delivery.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Yeruva Samrajya Lakshmi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Bhaskar C.
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Kishore Golla
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Anand K. Kondapi
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
- * E-mail:
| |
Collapse
|
25
|
Giacalone G, Hillaireau H, Fattal E. Improving bioavailability and biodistribution of anti-HIV chemotherapy. Eur J Pharm Sci 2015; 75:40-53. [PMID: 25937367 DOI: 10.1016/j.ejps.2015.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 04/07/2015] [Accepted: 04/13/2015] [Indexed: 11/25/2022]
Abstract
In the context of the treatment of HIV/AIDS, many improvements have been achieved since the introduction of the combination therapy (HAART). Nevertheless, no cure for this disease has been so far possible, because of some particular features of the therapies. Among them, two important ones have been selected and will be the subject of this review. The first main concern in the treatments is the poor drug bioavailability, resulting in repeated administrations and therefore a demanding compliance (drug regimens consist of multiple drugs daily intake, and non-adherence to therapy is among the important reasons for treatment failure). A second important challenge is the need to target the drugs into the so-called reservoirs and sanctuaries, i.e. cells or body compartments where drugs cannot penetrate or are distributed in sub-active concentrations. The lack of antiviral action in these regions allows the virus to lie latent and start to replicate at any moment after therapy suspension. Recent drug delivery strategies addressing these two limitations will be presented in this review. In the first part, strategies to improve the bioavailability are proposed in order to overcome the absorption or the target cell barrier, or to extend the efficacy time of drugs. In the second section, the biodistribution issues are considered in order to target the drugs into the reservoirs and the sanctuaries, in particular the mononuclear phagocyte system and the brain.
Collapse
Affiliation(s)
- Giovanna Giacalone
- Institut Galien Paris-Sud, Université Paris-Sud, Faculté de Pharmacie, 5 rue J.-B. Clément, F-92290 Châtenay-Malabry, France; CNRS, UMR 8612, F-92290 Châtenay-Malabry, France.
| | - Hervé Hillaireau
- Institut Galien Paris-Sud, Université Paris-Sud, Faculté de Pharmacie, 5 rue J.-B. Clément, F-92290 Châtenay-Malabry, France; CNRS, UMR 8612, F-92290 Châtenay-Malabry, France.
| | - Elias Fattal
- Institut Galien Paris-Sud, Université Paris-Sud, Faculté de Pharmacie, 5 rue J.-B. Clément, F-92290 Châtenay-Malabry, France; CNRS, UMR 8612, F-92290 Châtenay-Malabry, France.
| |
Collapse
|
26
|
Ramana LN, Anand AR, Sethuraman S, Krishnan UM. Targeting strategies for delivery of anti-HIV drugs. J Control Release 2014; 192:271-83. [PMID: 25119469 PMCID: PMC7114626 DOI: 10.1016/j.jconrel.2014.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 08/02/2014] [Accepted: 08/04/2014] [Indexed: 02/01/2023]
Abstract
Human Immunodeficiency Virus (HIV) infection remains a significant cause of mortality globally. Though antiretroviral therapy has significantly reduced AIDS-related morbidity and mortality, there are several drawbacks in the current therapy, including toxicity, drug–drug interactions, development of drug resistance, necessity for long-term drug therapy, poor bio-availability and lack of access to tissues and reservoirs. To circumvent these problems, recent anti-HIV therapeutic research has focused on improving drug delivery systems through drug delivery targeted specifically to host cells infected with HIV or could potentially get infected with HIV. In this regard, several surface molecules of both viral and host cell origin have been described in recent years, that would enable targeted drug delivery in HIV infection. In the present review, we provide a comprehensive overview of the need for novel drug delivery systems, and the successes and challenges in the identification of novel viral and host-cell molecules for the targeted drug delivery of anti-HIV drugs. Such targeted anti-retroviral drug delivery approaches could pave the way for effective treatment and eradication of HIV from the body.
Collapse
Affiliation(s)
- Lakshmi Narashimhan Ramana
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA University, Thanjavur, India; School of Chemical & Biotechnology, SASTRA University, Thanjavur, India
| | | | - Swaminathan Sethuraman
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA University, Thanjavur, India; School of Chemical & Biotechnology, SASTRA University, Thanjavur, India
| | - Uma Maheswari Krishnan
- Centre for Nanotechnology & Advanced Biomaterials, SASTRA University, Thanjavur, India; School of Chemical & Biotechnology, SASTRA University, Thanjavur, India.
| |
Collapse
|
27
|
Gupta R, Mehra NK, Jain NK. Development and characterization of sulfasalazine loaded fucosylated PPI dendrimer for the treatment of cytokine-induced liver damage. Eur J Pharm Biopharm 2013; 86:449-58. [PMID: 24189499 DOI: 10.1016/j.ejpb.2013.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/11/2013] [Accepted: 10/28/2013] [Indexed: 02/05/2023]
Abstract
The present investigation was aimed at exploring the targeting potential of sulfasalazine (NF-κB inhibitor drug) loaded fucose tethered poly (propylene imine) (PPI) dendritic nanoarchitecture (SSZ-FUCO-PPID) to Kupffer cells for effective management of cytokine-induced liver damage. The SSZ-FUCO-PPID formulation was characterized for entrapment efficiency, in vitro release, stability, toxicological investigations, macrophage uptake, NF-κB inhibition, and in vivo studies. In cell uptake assay the uptake of SSZ-FUCO-PPID was found to be higher and preferentially by J774 macrophage cell line. Cytokine assay suggested that the SSZ-FUCO-PPID potentially inhibited the IL-12 p40 production in LPS activated macrophages. Western blot analysis clearly suggested that SSZ-FUCO-PPID inhibited the activation of NF-κB as indicated by the absence of p-IκB band. Pharmacokinetic study revealed improved bioavailability, half-life and mean residence time of SSZ upon fucosylation of dendrimers. The biodistribution pattern clearly established the higher amount of SSZ-FUCO-PPID in liver. Hematological data suggest that the fucosylated formulations are less immunogenic as compared to unconjugated formulations. The results suggest that the SSZ-FUCO-PPID formulation holds targeting potential to Kupffer cells for the treatment of cytokine-induced liver damage.
Collapse
Affiliation(s)
- Richa Gupta
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour University, Sagar, India
| | - Neelesh Kumar Mehra
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour University, Sagar, India.
| | - Narendra Kumar Jain
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S. Gour University, Sagar, India.
| |
Collapse
|
28
|
Gupta R, Mehra NK, Jain NK. Fucosylated multiwalled carbon nanotubes for Kupffer cells targeting for the treatment of cytokine-induced liver damage. Pharm Res 2013; 31:322-34. [PMID: 24043294 DOI: 10.1007/s11095-013-1162-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/28/2013] [Indexed: 11/24/2022]
Abstract
PURPOSE To develop, characterize and exploring the sulfasalazine loaded fucoyslated multi walled carbon nanotubes for Kupffer cell targeting for effective management of cytokine-induce liver damage. METHODS Sulfasalazine was loaded into the fucosylated MWCNTs after subsequential functionalization (carboxylation, acylation and amidation) using dialysis membrane technique. The in vitro, in vivo studies were performed on macrophages J 774 cell line for Kupffer cells targeting for the treatment of cytokine-induced liver damage. RESULTS The loading of SSZ into SSZ-FUCO-MWCNTs was 87.77 ± 0.11% (n = 3). Sustained release was obtained from SSZ-FUCO-MWCNTs, with 89.12 ± 0.71% of SSZ released into medium at 48th hr. SSZ-FUCO-MWCNTs showed the 9.0 ± 0.23% hemolysis was drastically reduced from 21.62 ± 0.24% SSZMWCNTs 21.62 ± 0.24%. In SRB assay, SSZ-FUCO-MWCNTs showed more cytotoxicity than raw and SSZ-MWCNTs. In cytokine assay, SSZ- FUCO-MWCNTs exhibited significantly higher inhibition of IL-12 p40 secretion. In Western blot assay, SSZ-FUCO-MWCNTs significantly inhibit NF-κB activation. CONCLUSION The results suggested that the SSZ-FUCO-MWCNTs may be useful nano-carriers for targeted delivery to Kupffer cells in the treatment of cytokine-induced liver damage.
Collapse
Affiliation(s)
- Richa Gupta
- Pharmaceutics Research Laboratory Department of Pharmaceutical Sciences, Dr. H. S. Gour University, Sagar, MP, 470 003, India
| | | | | |
Collapse
|
29
|
Jain K, Kesharwani P, Gupta U, Jain NK. A review of glycosylated carriers for drug delivery. Biomaterials 2012; 33:4166-86. [DOI: 10.1016/j.biomaterials.2012.02.033] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 02/16/2012] [Indexed: 02/03/2023]
|
30
|
Dubey V, Nahar M, Mishra D, Mishra P, Jain NK. Surface structured liposomes for site specific delivery of an antiviral agent-indinavir. J Drug Target 2011; 19:258-269. [PMID: 20604740 DOI: 10.3109/1061186x.2010.499460] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The present investigation was aimed at targeting indinavir, a protease inhibitor to cells of mononuclear phagocyte system (MPS) via mannosylated liposomes. β-d-1-thiomannopyranoside residues were covalently coupled with dimyristoyl phosphatidylethanolamine (DMPE) to generate mannosylated-DMPE (Man-DMPE) conjugate which was further incorporated with disteroyl phosphatidyl choline and cholesterol to prepare mannosylated liposomes. The optimized mannosylated liposomes were nanometric in size (142 ± 2.8 nm) with optimum entrapment efficiency (88.7 ± 2.3%). Less than 20% cumulative drug release was observed from the prepared formulations in 24 h in phosphate buffer saline (pH 7.4). Cellular uptake studies performed on J774.A1 macrophage cell line via flow cytometric analysis depicted enhanced uptake of mannosylated liposomes as compared to plain liposomes. Annexin-V-fluorescein isothiocyanate/propidium iodide apoptosis assay delineated marginal cytotoxicity in macrophages from the developed formulation. Plasma and tissue distribution studies performed to assess the drug reach to macrophage rich regions depicted a significant level (P < 0.05) of indinavir in macrophage rich tissues like liver, spleen, and lungs from mannosylated liposomes as compared to plain liposomes and free drug. The conducted studies suggest the potential of indinavir loaded mannosylated liposomes for anti-human immunodeficiency virus therapy.
Collapse
Affiliation(s)
- Vaibhav Dubey
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. H. S.Gour University, Sagar, Madhya Pradesh, India
| | | | | | | | | |
Collapse
|
31
|
Targeted liposomal drug delivery to monocytes and macrophages. JOURNAL OF DRUG DELIVERY 2010; 2011:727241. [PMID: 21512579 PMCID: PMC3065850 DOI: 10.1155/2011/727241] [Citation(s) in RCA: 256] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 09/27/2010] [Indexed: 01/27/2023]
Abstract
As the role of monocytes and macrophages in a range of diseases is better understood, strategies to target these cell types are of growing importance both scientifically and therapeutically. As particulate carriers, liposomes naturally target cells of the mononuclear phagocytic system (MPS), particularly macrophages. Loading drugs into liposomes can therefore offer an efficient means of drug targeting to MPS cells. Physicochemical properties including size, charge and lipid composition can have a very significant effect on the efficiency with which liposomes target MPS cells. MPS cells express a range of receptors including scavenger receptors, integrins, mannose receptors and Fc-receptors that can be targeted by the addition of ligands to liposome surfaces. These ligands include peptides, antibodies and lectins and have the advantages of increasing target specificity and avoiding the need for cationic lipids to trigger intracellular delivery. The goal for targeting monocytes/macrophages using liposomes includes not only drug delivery but also potentially a role in cell ablation and cell activation for the treatment of conditions including cancer, atherosclerosis, HIV, and chronic inflammation.
Collapse
|
32
|
Zhang H, Ma Y, Sun XL. Recent developments in carbohydrate-decorated targeted drug/gene delivery. Med Res Rev 2010; 30:270-89. [PMID: 19626595 DOI: 10.1002/med.20171] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Targeted delivery of a drug or gene to its site of action has clear therapeutic advantages by maximizing its therapeutic efficiency and minimizing its systemic toxicity. Generally, targeted drug or gene delivery is performed by loading a macromolecular carrier with an appropriate drug or gene, and by targeting the drug/gene carrier to specific cell or tissue with the help of specific targeting ligand. The emergence of glycobiology, glycotechnology, and glycomics and their continual adaptation by pharmaceutical scientists have opened exciting avenue of medicinal applications of carbohydrates. Among them, the biocompatibility and specific receptor recognition ability confer the ability of carbohydrates as potential targeting ligands for targeted drug and gene delivery applications. This review summarizes recent progress of carbohydrate-decorated targeted drug/gene delivery applications.
Collapse
Affiliation(s)
- Hailong Zhang
- Department of Chemistry, Cleveland State University, Cleveland, Ohio 44115, USA
| | | | | |
Collapse
|
33
|
Choi JH, Lee YB. Nano-sized Drug Carriers and Key Factors for Lymphatic Delivery. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2010. [DOI: 10.4333/kps.2010.40.s.075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Gupta U, Jain NK. Non-polymeric nano-carriers in HIV/AIDS drug delivery and targeting. Adv Drug Deliv Rev 2010; 62:478-90. [PMID: 19913579 DOI: 10.1016/j.addr.2009.11.018] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 09/14/2009] [Indexed: 12/18/2022]
Abstract
Development of an effective drug delivery approach for the treatment of HIV/AIDS is a global challenge. The conventional drug delivery approaches including Highly Active Anti Retroviral Therapy (HAART) have increased the life span of the HIV/AIDS patient. However, the eradication of HIV is still not possible with these approaches due to some limitations. Emergence of polymeric and non-polymeric nanotechnological approaches can be opportunistic in this direction. Polymeric carriers like, dendrimers and nanoparticles have been reported for the targeting of anti HIV drugs. The synthetic pathways as well polymeric framework create some hurdles in their successful formulation development as well as in the possible drug delivery approaches. In the present article, we have discussed the general physiological aspects of the infection along with the relevance of non-polymeric nanocarriers like liposomes, solid lipid nanoparticles (SLN), ethosomes, etc. in the treatment of this disastrous disease.
Collapse
|
35
|
Sosnik A, Chiappetta DA, Carcaboso ÁM. Drug delivery systems in HIV pharmacotherapy: What has been done and the challenges standing ahead. J Control Release 2009; 138:2-15. [DOI: 10.1016/j.jconrel.2009.05.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2009] [Accepted: 05/04/2009] [Indexed: 01/04/2023]
|
36
|
Turánek J, Wang XF, Knötigová P, Koudelka Š, Dong LF, Vrublová E, Mahdavian E, Procházka L, Sangsura S, Vacek A, Salvatore BA, Neuzil J. Liposomal formulation of α-tocopheryl maleamide: In vitro and in vivo toxicological profile and anticancer effect against spontaneous breast carcinomas in mice. Toxicol Appl Pharmacol 2009; 237:249-57. [DOI: 10.1016/j.taap.2009.01.027] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 01/05/2009] [Accepted: 01/12/2009] [Indexed: 12/28/2022]
|
37
|
Kaur CD, Nahar M, Jain NK. Lymphatic targeting of zidovudine using surface-engineered liposomes. J Drug Target 2009; 16:798-805. [PMID: 19005941 DOI: 10.1080/10611860802475688] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The present investigation was aimed at lymphatic targeting of zidovudine (ZDV)-loaded surface-engineered liposomes (SE liposomes). Surface of liposomes was engineered by incorporation of charges (positive or negative) and site-specific ligand (mannose) in order to enhance localization to lymphatics, specifically to lymph node and spleen. Positively and negatively charged nanosized SE liposomes (120 +/- 10 nm) were prepared using stearylamine (SA) and dicetyl phosphate (DCP), respectively, while ligand-coated SE liposomes were prepared using mannose-terminated SA (mannose conjugate). The SE liposomes were characterized for shape and surface morphology, size, entrapment efficiency, and in vitro drug release. All the SE liposomes formulations showed biphasic ZDV release, whereas mannose-coated liposomes (MAN-Lip) significantly reduced (p < 0.05) drug release compared with conventional liposome (Lip). The organ distribution pattern of the SE liposomes exhibited significant reduction in free ZDV concentration in serum, whereas significantly increased quantity was detected in the spleen and lymph nodes (p < 0.05). Fluorescent microscopy suggested enhanced uptake and localization of the SE liposomes in the lymph nodes and spleen, which were in the order: mannose coated > negatively charged > positively charged > Lip. Thus, the SE liposomes appeared to be promising novel vesicular system for enhanced targeting of ZDV to lymphatics, in AIDS chemotherapy.
Collapse
Affiliation(s)
- Chanchal Deep Kaur
- Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Dr. Hari Singh Gour University, Sagar, India
| | | | | |
Collapse
|
38
|
Garg M, Garg BR, Jain S, Mishra P, Sharma RK, Mishra AK, Dutta T, Jain NK. Radiolabeling, pharmacoscintigraphic evaluation and antiretroviral efficacy of stavudine loaded 99mTc labeled galactosylated liposomes. Eur J Pharm Sci 2008; 33:271-81. [DOI: 10.1016/j.ejps.2007.12.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2007] [Revised: 11/05/2007] [Accepted: 12/13/2007] [Indexed: 10/22/2022]
|
39
|
Abstract
The design of well-defined particulate carrier systems with controlled size, shapes and physicochemical characteristics is becoming a focal point in the field of biomedicine and drug delivery. Dendrimers are one of the emerging technologies of recent times and have served as a unique platform to achieve the development as novel drug delivery scaffolds. Dendrimers may be engineered to meet the specific needs of biologically active agents, which can either be encapsulated within dendrimers or chemically attached to these units. The large number of active functional groups on the surface of dendrimers allows them to be meticulously tailored and to act as nano-scaffolds or nano-containers of various categories of drugs. The architecture of modified dendrimers has posed a challenge to drug delivery, in particular with respect to their in vivo metabolic fate. The drug delivery applications of dendrimers presented in this article provide an insight of their potential and substantiate the major roles for the future of these nanoconstructs.
Collapse
Affiliation(s)
- Narendra K Jain
- Dr Hari Singh Gour University, Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences, Sagar 4700 03, India.
| | | |
Collapse
|