View Article

Abstract

Due to a number of benefits, targeted medicine delivery has recently attracted increased interest. among the numerous avenues investigated for precise medication delivery. Particulate dispersions or solid particles with a size between 10 and 1000 nm are known as nanoparticles. A nanoparticle matrix is used to either dissolve, trap, encapsulate, or attach the medication. Depending on the preparation technique, one can produce nanoparticles, nanospheres, or nanocapsules. Controlling particle size, surface characteristics, and the release of pharmacologically active substances are the main objectives when designing nanoparticles as a delivery system in order to achieve the drug's site-specific activity at the therapeutically ideal pace and dosing regimen. The strategies for creating, characterizing, and using various nanoparticulate drug delivery systems are revealed in the current review.

Keywords

Nano particle, Drug delivery, Nanospheres, Nanocapsules

Introduction

The term "nanoparticle" refers to solid particles or particulate dispersions with sizes between 10 and 1000 nm. The medication was dissolved, trapped, enclosed, or joined to a nanoparticle matrix. [1,2]. More and more novel molecules are useful in the treatment of diseases. Numerous powerful medications have also been developed by biotechnology, although many of these drugs have delivery issues in biological systems [3,4]. Due to their incompatibilities and unique chemical makeup, their therapeutic efficiency is severely diminished [5,6]. The benefit of modern nanotechnology is that it makes it possible to distribute medicine at specified locations and times. The pharmaceutical business will be heavily impacted by the market for nanotechnology and medication delivery systems built on this technology. The number of goods and patents in this industry have dramatically increased in recent years [7,8]. The simplest application involves treating cancer, and there are various products on the market for this purpose, including Caelyx®, Doxil®, Trans drug®, and Abraxane®. The delivery of medicinal compounds to the desired place is a significant challenge in the management of many diseases. Traditional drug use is characterized by ineffectiveness, unfavorable side effects, poor biodistribution, and lack of selectivity [9,10]. These restrictions may be overcoming by methods like regulating medication distribution that deliver the drug to the site of action (Figure 1). Additionally, the medication delivery method offers defense against quick degradation or clearance. Additionally, it improves the medicine's absorption into the target tissues, requiring lower therapeutic doses. When there is a mismatch between a drug's dose or concentration and its therapeutic effects or harmful consequences, this sort of therapy is necessary.



       
            Picture1.png
       

    

Figure 1: Different types of drug delivery system


A more dependable method of medication delivery involves using specially developed carriers that are connected to drugs to target cells or particular tissues. This strategy is referred to as cell- or tissue-specific targeting. A more successful and essential method that serves as the foundation for nanotechnology is size reduction of targeted formulations and creating their pathways for appropriate drug delivery systems [11,12]. Recent developments in nanotechnology have demonstrated that nanoparticles have enormous potential as drug delivery systems. Different types of nanostructures with distinctive physicochemical and biological characteristics are produced using size reduction techniques and technologies [13,14]. 

Objectives

In order to accomplish the drug's site-specific effect at the therapeutically ideal rate and dose regimen, the main objectives of designing nanoparticles as a delivery system are to manage particle size, surface characteristics, and release of pharmacologically active substances. The goal of the medications is to have a precise action with minimal side effects and a better therapeutic index at a chosen site while avoiding undesired interactions at other sites. An example would be chemotherapy and enzyme replacement therapy for cancer [15].

Ideal Characteristics

  • A targeted medication delivery system should not be immunogenic or biochemically harmful. In vivo and in vitro, it is both physically and chemically stable.
  • Drug distribution should be limited to the intended cells, tissues, or organs and follow a uniform capillary distribution.
  • medication release that is predictable and under control.
  • Drug action is unaffected by drug release.
  • therapeutic dose of medication released.
  • very little drug leaking while in transit.
  • The utilised carriers must be easily removed from the body or biodegradable so there is no carrier-induced modification of the sick condition. The delivery method should be easy to prepare or at least somewhat simple reproductively and economically [15].

Many kinds of Nanoparticles

Although the kinds of nanoparticles listed below are all quite extensive and multifaceted, some of their fundamental characteristics and presently recognized applications in nanomedicine are presented here (Figure 2) [16,17].



       
            Picture2.png
       

    

Figure 2: Different types of nanoparticles


  1. Solid lipid nanoparticles (SLNs)
  2. Liposomes
  3. Nanostructured lipid carriers (NLC)
  4. Fullerenes
  5. Nanoshells
  6. Quantum dots (QD)
  7. Super paramagnetic nanoparticles
  1. Solid Lipid Nanoparticles (SLNs)

For colloid drug delivery applications, SLNs primarily consist of lipids that are in the solid phase at room temperature and emulsifying surfactants, whose mean diameters range from 50 nm to 1000 nm. SLNs are appealing due to their potential for improvement and offer special qualities such compact size, vast surface area, high drug loading, and the interaction of phases at the interfaces. Spray drying, high shear mixing, ultrasonication, and high-pressure homogenization (HPH) are common techniques for creating SLNs. Fatty acids (such as palmitic, decanoic, and behenic acids), triglycerides (such as trilaurin, trimyristin, and tripalmitin), steroids (such as cholesterol), partial glycerides (such as glyceryl monostearate and glyceryl behenate), and waxes (such as cetyl palmitate) are solid lipids used in SLN formulations. Soybean lecithin, phosphatidylcholine, poloxamer 188, sodium cholate, and sodium glycocholate are a few examples of the surfactants that are frequently used as emulsifiers to stabilize lipid dispersion. The use of physiological lipids in the synthesis of these solid lipid nanoparticles (SLN) has several benefits, including the avoidance of chemical solvents and a broad range of possible applications (intravenous, dermal and oral).

  1. Liposomes

The ejection of phospholipids results in the formation of liposomes, which are vesicular structures having an aqueous core encircled by a hydrophobic lipid bilayer. Because phospholipids are GRAS (generally acknowledged as safe) substances, the possibility of negative consequences is reduced. The hydrophobic bilayer prevents solutes, such as medicines, from passing through it; yet, hydrophobic molecules can be absorbed into the bilayer, allowing the liposome to transport both hydrophilic and hydrophobic molecules. Liposomes are valuable for medication delivery and cosmetic distribution applications because the lipid bilayer of these particles can fuse with other bilayers, such as the cell membrane, to facilitate the release of their contents. Nanoliposomes are another name for liposomes with vesicles that are in the nanometer range. Liposomes can range in size from 15 nm to several lm and comprise either a single layer of phospholipid bilayer membranes (unilamellar structure) or numerous layers of these membranes (multilamellar structure). Depending on their size range, unilamellar vesicles (ULVs) can be further divided into small unilamellar vesicles (SUVs) and large unilamellar vesicles (LUVs).

  1. Nanostructured Lipid Carriers (NLC)

Nanostructured Lipid Carriers are made from a mixture of solid and liquid lipids, however at body temperature, the particles are solid. Lipids are adaptable molecules that can build variously structured solid matrices, such as the lipid drug conjugate nanoparticles (LDC) and nanostructured lipid carriers (NLC), which have been developed to increase drug loading capacity. The solidified emulsion (dispersed phase) technologies are the foundation of NLC production. Due to drug ejection upon polymorphic change during storage, NLC may have an inadequate loading capacity, especially if the lipid matrix contains identical molecules. Drugs are released from lipid particles in the body by diffusion and concurrent lipid particle breakdown. In some circumstances, having a controlled quick release that goes beyond diffusion and degradation may be preferable. When the particles are delivered, an impulse should ideally cause this release to occur. Because of their highly disordered lipid structures, NLCs can accommodate the medication. By applying the trigger impulse to the matrix to transform it into a more ordered structure, a desired burst drug release can be started. This can cause the NLCs of specific structures to activate. NLCs can typically be used in situations where solid nanoparticles are more advantageous for medication delivery.

  1. Fullerenes

Any molecule that is made completely of carbon and has the shape of a hollow sphere, ellipsoid, or tube is referred to as a fullerene. Buck balls are another name for spherical fullerenes, whereas carbon nanotubes or buck tubes are another name for cylindrical fullerenes. Fullerenes have a structure that is similar to that of graphite, which is made up of stacked hexagonal ring sheets joined by pentagonal rings. They may also contain pentagonal (or occasionally heptagonal) rings, which could result in molecules that are porous. Endohedral fullerenes, which include the most prevalent fullerene, buckminsterfullerene, C60, are buckyball clusters or buck balls made up of fewer than 300 carbon atoms.

  1. Nano shells

Nanoshells, also known as core-shells, are tiny layers of another material that surround the spherical cores of specific compounds (concentric particles) and range in thickness from a few 1 to 20 nm. Compared to single component equivalents or nanoparticles of the same size, nanoshell particles are highly functional materials that exhibit changed and improved characteristics. You can alter their qualities by altering the constituent materials or the core-to-shell ratio. Semiconductors (dielectric materials like silica and polystyrene), metals, and insulators can all be used to create nanoshell materials. Due of their excellent stability, dielectric materials like silica and polystyrene are frequently employed as cores. Metal nanoshells are a brand-new class of composite spherical nanoparticles made of a thin metallic shell, usually made of gold, covering a dielectric core. Nanoshells have extremely advantageous optical and chemical properties for therapeutic and biomedical imaging uses. Other benefits that nanoshells have over typical organic dyes include enhanced optical qualities and a decreased propensity for chemical/thermal denaturation. Additionally, nanoshells can be easily changed using the same conjugation procedures that were utilized to link proteins to gold colloid. Nanoshells absorb heat and transfer it to the surrounding environment when a nanoshell and polymer matrix are lit with a resonant wavelength. This results in the network falling apart and the medication being released. In drug delivery systems based on core shell particles, the drug may either be encapsulated or adsorbed onto the shell surface. A specific functional group or an electrostatic stabilization technique is used by the shell to interact with the medication. It controls the medicine when it comes into contact with the biological system. With particular antibodies for tumors or damaged tissues, nanoshells can be marked for imaging purposes.

  1. Quantum Dots (QD)

The interface between various semiconductor materials can be found in the core shell nanocrystals of quantum dots, which are semiconductor nanocrystals. The diameter of quantum dots, which may be continually controlled between 2 and 10 nm, typically rises to 5 to 20 nm following polymer encapsulation. Renal filtration expeditiously removes particles smaller than 5 nm. For multiplexed, quantitative, and long-term fluorescence imaging and detection, semiconductor nanocrystals, which have special and intriguing optical features, have become a crucial tool in biomedical research. Small molecule hydrophobic drugs and imaging contrast agents can be encapsulated between the inorganic core and the amphiphilic polymer coating layer, with the QD core acting as the structural scaffold. Targeting biomolecules like antibodies, peptides, and aptamers can be immobilized onto the hydrophilic side of the amphiphilic polymer via covalent or non-covalent bonds, as can hydrophilic therapeutic agents like small interfering RNA (siRNA) and antisense oligodeoxynucleotide (ODN). This fully integrated nanostructure may behave like magic wands that not only recognize damaged cells but also bind to them and treat it. Detectable signals will also be emitted by it, allowing for real-time tracking of its course.

  1. Superparamagnetic Nanoparticles

The molecules known as super paramagnetic molecules are those that are drawn to a magnetic field but do not retain any magnetic properties once the field has been removed. For precise magnetic bio separations, iron oxide nanoparticles in the 5-100 nm range have been employed. For separation from the surrounding matrix, typical approaches involve coating the particles with antibodies to cell-specific antigens. Due to their paramagnetic characteristics, superparamagnetic nanoparticles have the ability to be seen in magnetic resonance imaging (MRI), guided to a specific place using a magnetic field, and heated to cause the release of a medication. Super paramagnetic nanoparticles fall within the category of inorganic-based particles with an iron oxide core that is covered in either inorganic substances (silica, gold), or organic substances (phospholipids, fatty acids, polysaccharides, peptides, or other surfactants and polymers). Superparamagnetic nanoparticles differ from ordinary nanoparticles in that they can be heated or guided to a specific place in the presence of an externally generated AC magnetic field due to their inducible magnetization. These qualities make them appealing for a variety of applications, including magnetically assisted cell transfection, drug delivery systems, magnetic hyperthermia (local heat source in the case of tumor therapy), and various separation techniques and contrast enhancing agents for MRI. Beads are already commercially available micron-sized polymer particles that contain SPIONs.

Application of Nanoparticle Technology


       
            Picture3.png
       

    
   Figure 3: Applications of nanoparticle technology


Cancer Therapy

The sort of therapy used to treat cancer patients today has saved the lives of many people, but the side effects of the treatment are severe and affect the entire body because the chemotherapeutic agents are non-specific. A biological process as complex as cancer might be thought of as an illness of many diseases. The rapid and uncontrolled division and multiplication of malignant cells is one of their distinguishing characteristics [18]. The fundamental goal of modern chemotherapy is to eliminate all cells that divide quickly. The drawback of this treatment is that it also kills off the body's other quickly multiplying cells, including those in the intestinal epithelium and hair follicles, leaving the patient to deal with potentially fatal side effects [19]. The creation of nanoparticles has opened up new possibilities for chemotherapy. Targeted medication delivery to the tumor site or to a specific subset of cells using cleverly constructed nanoparticles typically prevents adverse effects on surrounding healthy tissues and organs [20,21]. A number of systems have been tried out to deliver this kind of therapy. Another technique for the delivery of chemotherapeutic drugs is via micelles and liposomes. Additionally, because of its hydrophobic core and hydrophilic shell, micelles are a fantastic technique to make insoluble medications soluble. Further PEGylation of the micelle surface improves the ability of the nanocarriers to passively transport through the fenestrated vasculature of tumors and inflamed tissue, leading to increased drug concentration in tumors. The anticancer medications NK012, NK105, NK911, NC-6004, and SP1049C are among the polymeric micelles currently undergoing clinical trials [22]. One of these systems, Genexol-PM (paclitaxel), has been approved for the treatment of breast cancer patients. According to studies, dendrimers are highly branched macromolecules with numerous functional groups available for the attachment of drugs, targeting agents, and imaging agents. Their absorption, distribution, metabolism, and elimination (ADME) profiles are influenced by different structural features [23,24]. Methotrexate administration and effective localisation using folic acid, fluorescein, and a polyfunctional dendrimer method have all been documented [25]. By using biocompatible components and surface derivatization using PEGylation, acetylation, glycosylation, and different amino acids, nanoparticle treatments based on dendrimers can enhance the therapeutic index of cytotoxic medications [26,27]. Although there are various types of nanoparticles that have demonstrated potential in the treatment of cancer, carbon nanotubes are one of the newest systems. Single-walled carbon nanotubes (SWCNTs) and multiwalled carbon nanotubes (MWCNTs) are two types of carbon nanotubes (CNTs), an allotropic form of carbon with a cylindrical framework and deepening on number of sheets in concentric cylinders [28,29]. Since the hollow core of carbon nanotubes is extremely hydrophobic, it is simple to load them with medications that are insoluble in water. Due to the wide surface area, it is possible to tailor the outer surface for a specific cancer receptor as well as contrast chemicals [30]. The spherical molecule Buckminsterfullerene C60 and its derivatives are lastly investigated for the treatment of cancer [31,32]. In addition to being a great scavenger of reactive oxygen species (ROS), fullerene C60 may bind up to six electrons.   According to a study, fullerene nanocrystals (Nano-C60) can increase the cytotoxicity of chemotherapy drugs. As a result, adjunct chemotherapy using Nano-C60 can be further investigated [33,34]. In a subsequent investigation, Fullerene C60 and Doxorubicin were combined, and found that the tumor volumes of the treated rats (C60 + Dox) were 1.4 times lower than those of the untreated rats in the control group [33]. Additionally, it is believed that the C60 + Dox complex exerts its antitumor effects both directly on tumor cells and indirectly through immunomodulation. Diagnostic tests, clause 5.2 Although it is not now feasible for clinical usage, the use of nanoparticles for diagnostic purposes has received significant academic attention [35]. Fluorescent nanoparticles offer researchers a solution to address these drawbacks since current diagnostic testing technology is hampered by the shortcomings of fluorescent markers, including fading of fluorescence after single use, color matching, and restricted use of dyes due to a bleeding effect [36]. The discovery of quantum dots, which can be produced on demand in a wide variety of clearly distinct hues, was a significant advance. Their high quantum yield, tunable emission spectrum, and photostability are all features of their wide absorption spectrum, which extends from ultraviolet to a visible spectrum wavelength. Where a particular particle lands in the spectrum depends on the size of the nanodot. Longer wavelengths and narrower emission are characteristic of larger particles [37-39]. Quantum dot tagging has a number of benefits. They are initially easily excited by white light. Second, they can be connected to biomolecules that can stay inside a biological organism for a considerable amount of time to investigate different biomechanics. By labeling diverse biological components with nanodots of a particular hue, this technology also enables the simultaneous monitoring of numerous biological events [40]. Theranostic nanoparticles, which can be utilized for both diagnosis and treatment, have attracted a lot of attention recently [41].   Many kinds of nanoparticles, including drug conjugates, dendrimers, surfactant aggregates (micelles and vesicles), core-shell particles, and carbon nanotubes, have successfully used this method. It is possible to track the pathway and localization of these nanoparticles at the target site as well as drug action to evaluate therapeutic response by combining both the drug and imaging agent in one clever formulation [42].

Nanoparticles for Lymph Targeting

Lymph targeting is primarily used to deliver an efficient anticancer treatment and stop the metastasis of tumor cells by building up the drug in the local lymph node through subcutaneous delivery. Lymph node targeting also aims to improve the oral bioavailability of macromolecular drugs like polypeptides and proteins, which are absorbed via Peyer's patches in the intestine, as well as the localization of diagnostic agents to the regional lymph node for the visualization of lymphatic vessels prior to surgery [43]. Numerous researches have been conducted so far for the lymphatic targeting employing medication carriers in nanoparticles [44]:

    1. Anticancer drug-loaded polyalkylcyanoacrylate nanoparticles for peritoneal cavity tumors.
    2. Insulin-loaded polyisobutylcyanoacrylate nanoparticles for peyer's patch-based peroral peptide administration.
    3. Poly (lactide-co-glycolide) nanoparticles for the lymphatic transport of diagnostic chemicals.
    4. Magnetic resonance imaging using magnetite-dextran nanoparticles as a contrast agent. 
    5. Marker-bearing Polyalkylcyanoacrylate Nanocapsules for lymphatic distribution (ASA) [45].

HIV and AIDS Treatment

Human immunodeficiency virus (HIV) infection can cause acquired immune deficiency syndrome (AIDS), a fatal illness when a person's immune system is nearly wiped out [46]. First therapy was painstakingly complex, requiring the majority of patients to take 30–40 tablets every day. Therapeutic advances over the past ten years have allowed for a daily pill count of just a handful [47]. The development of polymeric nanoparticles that carry antiretroviral (ARV) medications intracellularly as well as to the brain has been proven to increase the efficacy of this treatment [48]. In order to prevent HIV infections, this technology can also be utilized in conjunction with immunizations [49]. Depending on the stages of the HIV life cycle that they respond best to, antiretroviral medications used to treat HIV can be divided into different categories. Highly effective antiretroviral therapy (HAART), a combination of several medications (three or more), is used to avoid the emergence of resistance and to actively halt the progression of HIV [50]. Antiretroviral medicine delivery and compliance have greatly benefited from the use of nanotechnology. When administered orally or through other non-parental means (suppository and patches, etc.), antiretroviral medications must be able to pass the mucosal epithelial barrier. HIV mostly infects and thrives in lymphoid tissues. According to several studies [51,52]. Antiretroviral drug-loaded nanoparticles can successfully target monocytes and macrophages in vitro. Some study provided an excellent illustration of the superiority and effectiveness of nanoparticle systems for sustained and targeted medication administration. The researchers created nanoparticles encasing the antiretroviral medications ritonavir, lopinavir, and efavirenz using poly (lactic-co-glycolic acid) (PLGA). While free medicines were removed within 48 hours (2 days), the nanoparticle method produced sustained drug release for more than 4 weeks (28 days). According to study, the central nervous system (CNS) is another location where HIV can infect and thrive, leading to the devastating HIV-associated neurocognitive disorder (HAND). Many studies have successfully delivered anti-HIV drugs using nanoparticles that can breach the blood-brain barrier (BBB) by endocytosis or phagocytosis [53-55].  

Nutraceutical Delivery

Nutraceuticals are standardized, food-derived ingredients with observable health advantages. They are frequently ingested in addition to different allopathic treatments in order to offer additional health advantages and lower the chance of developing a number of chronic illnesses [56]. Similar to the situation with any other medicine, food matrix interactions, water solubility, degradation/metabolism, and epithelial permeability all have an impact on the bioavailability and consequently effectiveness of orally taken nutraceuticals. The majority of nutraceuticals, including polyunsaturated lipids, various phytochemicals, and fat-soluble vitamins (A, D, E, and K), are lipophilic molecules. Again, nanotechnology provides all-encompassing support, and the majority of studies have focused on enhancing the solubility processes of nutraceuticals through formulations with nanoparticles [57,58]. Numerous nutraceuticals have anti-inflammatory, antioxidant, antiapoptotic, and antiangiogenic properties; curcumin (diferuloylmethane) is the most well-known and extensively researched of them. Numerous strategies have been used to solve this problem, including liposomes, phospholipid vesicles, and polymer-based nano-formulation [59,60]. It is nearly water-insoluble and has very poor bioavailability. In comparison to curcumin co-administered with piperine (an absorption enhancer), curcumin's oral bioavailability was found to be 9 times higher [61]. When compared to curcumin powder, another study utilizing colloidal nanoparticles of curcumin called Theracurmin shown inhibitory effects against alcohol intoxication and an area under the curve (AUC) that was 40 times higher in rats and 27 times higher in healthy human volunteers [62]. Several plants naturally contain resveratrol, a significant non-flavonoid polyphenol, however it is most prevalent in Vitis vinifera, labrusca, and muscadine grapes [63]. According to study, it exhibits antioxidant, cardioprotective, anti-inflammatory, and anticancer properties [64]. Although resveratrol has a low solubility and a fair amount of bioavailability, it is quickly metabolized and excreted from the body [65,66]. The more prevalent and biologically active of the two geometric isomers of resveratrol, trans-resveratrol, is photosensitive and transforms into cis-resveratrol in the presence of light [67]. There have been numerous reports of resveratrol nanoformulations that increase the pharmacokinetic profile and bioavailability [68]. Zein-based nanoparticles, nanoemulsions, liposomes, cyclodextrins, and dual nanoencapsulation methods [69-71]. Recently, the blood-brain barrier was used to test the neuroprotective benefits of resveratrol using solid lipid nanoparticles coated with apolipoprotein E for LDL receptor recognition [72].

Nanoparticles for Brain Delivery

One of the obstacles for medications like antibiotics, antineoplastics, and certain neuroleptics is the blood-brain barrier. Drug delivery to the brain using nanoparticles has been suggested as one way to get over this obstacle. The hexapeptide dalargin, the dipeptide kyotropin, loperamide, tubocurarine, and doxorubicin are among the medications that have been effectively employed for brain targeting with nanoparticles. There are several possibilities for improved drug delivery to the brain utilizing nanoparticles:

  1. A greater concentration gradient at the blood–brain barrier, which might improve transport through the endothelial cell layer and, as a result, boost retention in the brain.
  2. Lipids in the endothelial cell membrane are dissolved by the surfactant activity of nanoparticles, resulting in fluidization of the membrane and improved drug permeability to BBB.
  3. The permeability of drugs or drug-nanoparticle conjugates through these channels is increased due to the loosening of tight junctions between endothelial cells.
  4. After the endothelial cells endocytoze nanoparticles, the medication is released intracellularly.
  5. Drug-bound nanoparticle transcytosis via the endothelial cell layer [73].

Nanoparticles have been recommended for brain administration by Kreuter and colleagues in a number of studies described employing poly (butyl cyanoacrylate) nanoparticles coated with polysorbate 80 to transport the hexapeptide dalargin across the blood-brain barrier [74]. When compared to all other controls, including a straightforward mixture of the three components (drugs, nanoparticles, and surfactant) mixed right before intravenous injection, the intravenous injection of polysorbate 80 coated nanoparticles with sorbed drug produced a significant analgesic effect in mice. According to fluorescent and electron microscopy examinations, the endothelial cells in the blood vessels of the brain phagocytosed the d polysorbate 80-coated nanoparticles, which allowed the drug to flow through the particle-bound drug. Using nanoparticle technology, several neuropeptides are also transported across the blood-brain barrier. Neuropeptides such leu-enkephalin dalargin and met-enkephalin kyotorphin typically do not cross the Blood-Brain Barrier (BBB) when administered systemically. These neuropeptides were adsorbed onto poly (butyl cyanoacrylate) nanoparticles and coated with polysorbate 80 to allow them to pass across the BBB [73].

Nanoparticles for Ocular Delivery

The majority of uses for ocular drug delivery systems are in the treatment of glaucoma, particularly with cholinergic agonists like pilocarpine. Utilizing nanoparticles with biodegradable qualities, the short elimination half-life of aqueous eye drops (likely caused by lachrymal drainage) can be increased from a very brief time (1-3 min) to a longer duration (15 min). These include polyester nanoparticles, polyalkylcyanoacrylate nanoparticles and albumin. Additionally, it has been shown that nanoparticles stick to inflamed tissue more firmly than they do to healthy tissue; as a result, these particles could be utilized to direct anti-inflammatory medications onto inflamed eyes. The polyalkylcyanoacrylate nanoparticles, in particular PHCA nanoparticles, are touted for a number of benefits, such as their biodegradability, tissue adherence, and increased elimination half-life of their drainage paired with a delayed clearance. Polyalkylcyanoacrylate nanoparticles that were pilocarpine and betaxolol loaded were shown to be able to extend and maintain the decreased intraocular pressure in rabbits for more than 9 hours. Pilocarpine-loaded polybutylcyanoacrylate nanoparticles were employed for ocular administration by Zimmer in 1994. In terms of reducing intraocular pressure (IOP) and enhancing mitosis, pilocarpine's pharmacokinetics and pharmacodynamic response were improved. The traditional method of coating pilocarpine-loaded nanoparticles with bioadhesive or viscous polymers has the potential to boost their effectiveness. The bio adhesive qualities of the polymers methylcellulose, polyvinyl alcohol, hydroxyl propylmethylcellulose, and carbopol 941 led to their selection [75].

Antibody Targeting of Nanoparticles

Numerous studies have shown the use of nanoparticles mediated by antibodies to create tailored drug delivery systems, particularly in the context of cancer treatment. The therapeutic effectiveness of a medicinal substance can be enhanced by targeting antibodies, which can also enhance drug distribution and concentration at the intended site of action. In order ma cells, McCarron et al. examined two innovative techniques. To target nanoparticles, they employed CD95/APO-1 antibody and poly (lactide) polyto produce immunonanoparticles with increased therapeutic impact against colorectal carcinomers. Dendrimer-magnetic nanoparticles were employed by Pan et al. to deliver gene-targeted cancer therapies effectively. The utilization of nanostructured calcium nanophosphates for non-viral gene delivery has been documented by Olton et al. They have also investigated how synthesis factors affect transfection effectiveness [76].

Nanoparticles for DNA Delivery

Recently, nanoparticles have been utilized to demonstrate their stability in the bio-environment and as a delivery vehicle for the transfection of plasmid DNA. In 1998, Truong-Le and colleagues created a unique gene delivery technology based on the utilization of DNA-gelatin nanoparticles (Nano spheres) produced by the complicated concertation of gelatin and plasmid A caused by salt. Compared to naked DNA, nanosphere-DNA that had been treated in bovine serum was more resistant to nuclease digestion. The interaction of different bioactive agents with the components of the matrix, physical entrapment, or covalent conjugation might all result in the bioactive agents being enclosed in the nanospheres. Chloroquine and calcium-containing DNA gelatin nanoparticles were further developed by Troung-Le and colleagues in 1999. As a means of delivering genes, the targeting ligand, transferring, was covalently attached to the gelatin. The presence of calcium and Nano spheres containing transferring DNA were necessary for optimal cell transfection by this method. Using either chitosan-DNA complexes or chitosan-DNA nanospheres, James and colleagues developed chitosan-DNA hybrid colloidal systems in 2001 and found comparatively improved gene expression [77].

Nanoparticles in Diagnostic Medicine

A highly intriguing paper on the use of nanoparticles in diagnostic medicine has been published. They employed hydrophilic, magnetic nanocrystals with antibody conjugates as smart nano probes for MRI's ultrasensitive breast cancer detection. Thermal breakdown was used to create the MnFe2 O4 nanocrystals used as MRI contrast agents. Amphiphilic triblock copolymers were then used to modify the surfaces. They clearly shown their superiority as a contrast medium for locating breast cancer lesions. By connecting a molecule to dielectric particles with a rare earth oxide core and a polysiloxane shell containing fluorescein for bio detection. Study have demonstrated various approaches to detect streptavidin. Gadolinium hydroxide and dysprosium oxide are members of a novel and intriguing family of magnetic nanoparticles that have been studied using X-ray diffraction, NMR relaxometry, and magnetometry in various fields. These are excellent for use in diagnostic procedures. For the purpose of finding circulating breast cancer cells, Shao et al. employed nanotube antibody biosensor arrays. The unique method for detecting cancer cells utilizing a nanotube and antibody combination is described in this research for the first time [77].

CONCLUSION

The commercial application of nanotechnology is quite promising. Many believe that the use of nanotechnology in medication delivery will lead to the development of innovative therapies that could completely alter the pharmaceutical and biotechnology sectors. There are several areas of interest where there will be efficient and safer tailored treatments for a variety of clinical applications, and numerous nanotechnology platforms are being researched, either in development or in clinical stages. It will soon be evolving for the benefit of all humankind.

REFERENCES:

  1. Francisco JG, Ma LM, Paloma G, Ruth R. Nanotechnology and food industry. In:. In:Tech Dr. Valdez B, editor. Scientific, health and social aspects of the food industry. ISBN: 978-953-307-916-5; 2012. p. 1-35.
  2. Michele F, Oliveira, Pedro PG, Guimarães, Alinne DM, Gomes et al. Strategies to target tumors.
  3. Mohanraj VJ, Chen Y. Nanoparticles - a review. Trop J Pharm Res. 2006;5(1):561-73. doi: 10.4314/tjpr.v5i1.14634.
  4. Hoet PHM, Hohlfeld IB, Salata OV. Nanoparticles -known and unknown health risks. J Nano Biotechnol. 2004;02:01-15.
  5. Thassu YPD. Drug delivery nanoparticles formulation and characterization. Drugs Pharm Sci. 2007;191.
  6. Adams M. The top ten technologies: where is nanotechnology? 2004;2004 [cited 2013 7/05/2013]; about scope & presence of Nanotechnology.
  7. Badjatya JK. Enhancement of Dissolution of Poorly water Soluble aceclofenac by preparing Nanoparticles Inventi Rapid. NDDS. 2012;2012:01-2.
  8. Mohmmed Ali DAAS, Showkat R, Wadood Siddiqui A. GPAT: A Ideal book for Pharmacy Competative Exams. 2nd ed GPAT: A Ideal; 2012.
  9. Moghimi SM, Kissel T. Particulate nanomedicines?. Adv Drug Deliv. 2006;58(14):1451-5. doi: 10.1016/j.addr.2006.09.010.
  10. Roger A. Nanotechnology applications for drug delivery. Drug Deliv Nanotechnol. 2004:21-8.
  11. des Rieux AD, Fievez V, Garinot M, Schneider YJ, Préat V. Nanoparticles as potential oral delivery systems of proteins and vaccines: A mechanistic approach. J Control Release. 2006;116(1):1-27. doi: 10.1016/j.jconrel.2006.08.013, PMID 17050027.
  12. Storm G, Crommelin DJA. Liposomes: quo vadis? Pharm Sci Technol Today. 1998;1(1):19-31. doi: 10.1016/S1461-5347(98)00007-8.
  13. Couvreur P, Vauthier C. Nanotechnology: intelligent design to treat complex diseases. Pharm Res. 2006;23(7):1417-50. doi: 10.1007/s11095-006-0284-8, PMID 16779701.
  14. Jain S, Shukla K, Jain V, Saraf S. Nanoparticles: emerging carriers for delivery of bioactive agents. Pharma Times; 2006. In: 39:30-5.
  15. Phillipos T. Nanoparticles safe! About .co [guide]; 2009.
  16. Michele F, Oliveira, Pedro PG, Guimarães, Alinne DM, Gomes et al. Strategies to target tumors.
  17. Rick D. Berkeley considering need for nano safety.articles.sfgate.com; 2006.
  18. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74. doi: 10.1016/j.cell.2011.02.013, PMID 21376230.
  19. Baudino TA. Targeted cancer therapy: the next generation of cancer treatment. Curr Drug Discov Technol. 2015;12(1):3-20. doi: 10.2174/1570163812666150602144310, PMID 26033233.
  20. Shen B, Ma Y, Yu S, Ji C. Smart multifunctional magnetic nanoparticle-based drug delivery system for cancer thermo-chemotherapy and intracellular imaging. ACS Appl Mater Interfaces. 2016;8(37):24502-8. doi: 10.1021/acsami.6b09772, PMID 27573061.
  21. Huang B, Abraham WD, Zheng Y, Bustamante López SC, Luo SS, Irvine DJ. Active targeting of chemotherapy to disseminated tumors using nanoparticle-carrying T cells. Sci Transl Med. 2015;7(291):291ra94. doi: 10.1126/scitranslmed.aaa5447, PMID 26062846.
  22. Oerlemans C, Bult W, Bos M, Storm G, Nijsen JF, Hennink WE. Polymeric micelles in anticancer therapy: targeting, imaging and triggered release. Pharm Res. 2010;27(12):2569-89. doi: 10.1007/s11095-010-0233-4, PMID 20725771.
  23. Somani S, Dufès C. Applications of dendrimers for brain delivery and cancer therapy. Nanomedicine (Lond). 2014;9(15):2403-14. doi: 10.2217/nnm.14.130, PMID 25413857.
  24. Kaminskas LM, Boyd BJ, Porter CJ. Dendrimer pharmacokinetics: the effect of size, structure and surface characteristics on ADME properties. Nanomedicine (Lond). 2011;6(6):1063-84. doi: 10.2217/nnm.11.67, PMID 21955077.
  25. Quintana A, Raczka E, Piehler L, Lee I, Myc A, Majoros I, et al. Design and function of a dendrimerbased therapeutic nanodevice targeted to tumor cells through the folate receptor. Pharm Res. 2002;19(9):1310-6. doi: 10.1023/a:1020398624602, PMID 12403067.
  26. Baker Jr. JR. Dendrimer-based nanoparticles for cancer therapy. Hematology Am Soc Hematol Educ Program. 2009:708-19. doi: 10.1182/asheducation-2009.1.708, PMID 20008257.
  27. Cheng Y, Zhao L, Li Y, Xu T. Design of biocompatible dendrimers for cancer diagnosis and therapy: current status and future perspectives. Chem Soc Rev. 2011;40(5):2673-703. doi: 10.1039/c0cs00097c, PMID 21286593.
  28. Rastogi V, Yadav P, Bhattacharya SS, Mishra AK, Verma N, Verma A, et al. Carbon nanotubes: an emerging drug carrier for targeting cancer cells. J Drug Deliv. 2014;2014:670815. doi: 10.1155/2014/670815, PMID 24872894.
  29. Sanginario A, Miccoli B, Demarchi D. Carbon nanotubes as an effective opportunity for cancer diagnosis and treatment. Biosensors. 2017;7(1):9. doi: 10.3390/bios7010009, PMID 28212271.
  30. Dinesh B, Bianco A, Ménard-Moyon C. Designing multimodal carbon nanotubes by covalent multi-functionalization. Nanoscale. 2016;8(44):18596-611. doi: 10.1039/c6nr06728j, PMID 27805213.
  31. Murugesan S, Mousa SA, O’connor LJ, Lincoln DW, Linhardt RJ. Carbon inhibits vascular endothelial growth factor- and fibroblast growth factor-promoted angiogenesis. FEBS Lett. 2007;581(6):1157-60. doi: 10.1016/j.febslet.2007.02.022, PMID 17331505.
  32. Chen Z, Mao R, Liu Y. Fullerenes for cancer diagnosis and therapy: preparation, biological and clinical perspectives. Curr Drug Metab. 2012;13(8):1035-45. doi: 10.2174/138920012802850128, PMID 22380017.
  33. Prylutska SV, Skivka LM, Didenko GV, Prylutskyy YI, Evstigneev MP, Potebnya GP, et al. Complex of C60 fullerene with doxorubicin as a promising agent in antitumor therapy. Nanoscale Res Lett. 2015;10(1):499. doi: 10.1186/s11671-015-1206-7, PMID 26714861.
  34. Zhang Q, Yang W, Man N, Zheng F, Shen Y, Sun K, et al. Autophagy-mediated chemosensitization in cancer cells by fullerene C60 nanocrystal. Autophagy. 2009;5(8):1107-17. doi: 10.4161/auto.5.8.9842, PMID 19786831.
  35. Kolluru LP, Rizvi SAA, D’Souza M, D’Souza MJ. Formulation development of albumin based theragnostic nanoparticles as a potential tumor targeting and delivery system. J Drug Target. 2013;21(1):77-86. doi: 10.3109/1061186X.2012.729214, PMID 23036042.
  36. Wolfbeis OS. An overview of nanoparticles commonly used in fluorescent bioimaging. Chem Soc Rev. 2015;44(14):4743-68. doi: 10.1039/c4cs00392f, PMID 25620543.
  37. Emerich DF, Thanos CG. Targeted nanoparticle-based drug delivery and diagnosis. J Drug Target. 2007;15(3):163-83. doi: 10.1080/10611860701231810, PMID 17454354.
  38. Li J, Zhu JJ. Quantum dots for fluorescent biosensing and bio-imaging applications. Analyst. 2013;138(9):2506-15. doi: 10.1039/c3an36705c, PMID 23518695.
  39. Michalet X, Pinaud FF, Bentolila LA, Tsay JM, Doose S, Li JJ, et al. Quantum dots for live cells, in vivo imaging, and diagnostics. Science. 2005;307(5709):538-44. doi: 10.1126/science.1104274, PMID 15681376.
  40. Datta R, Jaitawat SS. Nanotechnology - the new frontier of medicine. Med J Armed Forces India. 2006;62(3):263-8. doi: 10.1016/S0377-1237(06)80016-X, PMID 27365690.
  41. Janib SM, Moses AS, MacKay JA. Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliv Rev. 2010;62(11):1052-63. doi: 10.1016/j.addr.2010.08.004, PMID 20709124.
  42. Bhojani MS, Van Dort M, Rehemtulla A, Ross BD. Targeted imaging and therapy of brain cancer using theranostic nanoparticles. Mol Pharm. 2010;7(6):1921-9. doi: 10.1021/mp100298r, PMID 20964352.
  43. Subrahmanyam CVS. Textbook of Physical Pharmaceutics (Thorouly revised & Enlarged). 2’nd Reprint (August) ed. Textbook of Physical Pharmaceutics. 1 Delhi: Vallabh Prakashan. 2005;388.
  44. Couvreur P, Vauthier C. Nanotechnology: intelligent design to treat complex diseases. Pharm Res. 2006;23(7):1417-50. doi: 10.1007/s11095-006-0284-8, PMID 16779701.
  45. Raval AJ, Madhabahi MP. Preparation and characterization of nanoparticles for solubility and dissolution rate enhancement of meloxicam. Int J Pharm. 2011;01:42-9.
  46. Moss JA. HIV/AIDS review. Radiol Technol. 2013;84(3):247-67; quiz p.268. PMID 23322863.
  47. Bartlett JG, Moore RD. Improving HIV therapy. Sci Am. 1998;279(1):84-7, 89. doi: 10.1038/scientificamerican0798-84, PMID 9648300.
  48. Mamo T, Moseman EA, Kolishetti N, Salvador-Morales C, Shi J, Kuritzkes DR, et al. Emerging nanotechnology approaches for HIV/AIDS treatment and prevention. Nanomedicine (Lond). 2010;5(2):269-85. doi: 10.2217/nnm.10.1, PMID 20148638.
  49. Khalil NM, Carraro E, Cótica LF, Mainardes RM. Potential of polymeric nanoparticles in AIDS treatment and prevention. Expert Opin Drug Deliv. 2011;8(1):95-112. doi: 10.1517/17425247.2011.543673, PMID 21143001.
  50. Crabtree-Ramírez B, Villasís-Keever A, Galindo-Fraga A, del Río C, Sierra-Madero J. Effectiveness of highly active antiretroviral therapy (HAART) among HIV-infected patients in Mexico. AIDS Res Hum Retroviruses. 2010;26(4):373-8. doi: 10.1089/aid.2009.0077, PMID 20377418.
  51. Shah LK, Amiji MM. Intracellular delivery of saquinavir in biodegradable polymeric nanoparticles for HIV/AIDS. Pharm Res. 2006;23(11):2638-45. doi: 10.1007/s11095-006-9101-7, PMID 16969696.
  52. Mallipeddi R, Rohan LC. Progress in antiretroviral drug delivery using nanotechnology. Int J Nanomedicine. 2010;5:533-47. PMID 20957115.
  53. Spudich SS, Ances BM. Central nervous system complications of HIV infection. Top Antivir Med. 2011;19(2):48-57. PMID 21868822.
  54. Rao KS, Ghorpade A, Labhasetwar V. Targeting anti-HIV drugs to the CNS. Expert Opin Drug Deliv. 2009;6(8):771-84. doi: 10.1517/17425240903081705, PMID 19566446.
  55. Nowacek AS, McMillan J, Miller R, Anderson A, Rabinow B, Gendelman HE. Nanoformulated antiretroviral drug combinations extend drug release and antiretroviral responses in HIV-1- infected macrophages: implications for neuroAIDS therapeutics. J Neuroimmune Pharmacol. 2010;5(4):592-601. doi: 10.1007/s11481-010-9198-7, PMID 20237859.
  56. Aggarwal BB, Van Kuiken ME, Iyer LH, Harikumar KB, Sung B. Molecular targets of nutraceuticals derived from dietary spices: potential role in suppression of inflammation and tumorigenesis. Exp Biol Med (Maywood). 2009;234(8):825-49. doi: 10.3181/0902-MR-78, PMID 19491364.
  57. Acosta E. Bioavailability of nanoparticles in nutrient and nutraceutical delivery. Curr Opin Colloid Interface Sci. 2009;14(1):3-15. doi: 10.1016/j.cocis.2008.01.002.
  58. McClements DJ, Li F, Xiao H. The nutraceutical bioavailability classification scheme: classifying nutraceuticals according to factors limiting their oral bioavailability. Annu Rev Food Sci Technol. 2015;6:299-327. doi: 10.1146/annurev-food-032814-014043, PMID 25705933.
  59. Mohanty C, Sahoo SK. The in vitro stability and in vivo pharmacokinetics of curcumin prepared as an aqueous nanoparticulate formulation. Biomaterials. 2010;31(25):6597-611. doi: 10.1016/j.biomaterials.2010.04.062, PMID 20553984.
  60. Carvalho DdM, Takeuchi KP, Geraldine RM, Moura CJd, Torres MCL. Production, solubility and antioxidant activity of curcumin nanosuspension. Food Sci Technol (Campinas). 2015;35(1):115-9. doi: 10.1590/1678-457X.6515.
  61. Shaikh J, Ankola DD, Beniwal V, Singh D, Kumar MN. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur J Pharm Sci. 2009;37(3-4):223-30. doi: 10.1016/j.ejps.2009.02.019, PMID 19491009.
  62. Sasaki H, Sunagawa Y, Takahashi K, Imaizumi A, Fukuda H, Hashimoto T, et al. Innovative preparation of curcumin for improved oral bioavailability. Biol Pharm Bull. 2011;34(5):660-5. doi: 10.1248/bpb.34.660, PMID 21532153.
  63. Celotti E, Ferrarini R, Zironi R, Conte LS. Resveratrol content of some wines obtained from dried Valpolicella grapes: recioto and Amarone. J Chromatogr A. 1996;730(1-2):47-52. doi: 10.1016/0021-9673(95)00962-0, PMID 8680595.
  64. Summerlin N, Soo E, Thakur S, Qu Z, Jambhrunkar S, Popat A. Resveratrol nanoformulations: challenges and opportunities. Int J Pharm. 2015;479(2):282-90. doi: 10.1016/j.ijpharm.2015.01.003, PMID 25572692.
  65. Walle T. Bioavailability of resveratrol. Ann N Y Acad Sci. 2011;1215:9-15. doi: 10.1111/j.1749-6632.2010.05842.x, PMID 21261636.
  66. Kapetanovic IM, Muzzio M, Huang Z, Thompson TN, McCormick DL. Pharmacokinetics, oral bioavailability, and metabolic profile of resveratrol and its dimethylether analog, pterostilbene, in rats. Cancer Chemother Pharmacol. 2011;68(3):593-601. doi: 10.1007/s00280-010-1525-4, PMID 21116625.
  67. Trela BC, Waterhouse AL. Resveratrol: isomeric molar absorptivities and stability. J Agric Food Chem. 1996;44(5):1253-7. doi: 10.1021/jf9504576.
  68. Sanna V, Siddiqui IA, Sechi M, Mukhtar H. Resveratrol-loaded nanoparticles based on poly(epsilon-caprolactone) and poly (D, L-lactic-co-glycolic acid)-poly (ethylene glycol) blend for prostate cancer treatment. Mol Pharm. 2013;10(10):3871-81. doi: 10.1021/mp400342f, PMID 23968375.
  69. Penalva R, Esparza I, Larraneta E, González-Navarro CJ, Gamazo C, Irache JM. Zein-based nanoparticles improve the oral bioavailability of resveratrol and its anti-inflammatory effects in a mouse model of endotoxic shock. J Agric Food Chem. 2015;63(23):5603-11. doi: 10.1021/jf505694e, PMID 26027429.
  70. Catania A, Barrajón-Catalán E, Nicolosi S, Cicirata F, Micol V. Immunoliposome encapsulation increases cytotoxic activity and selectivity of curcumin and resveratrol against HER2 overexpressing human breast cancer cells. Breast Cancer Res Treat. 2013;141(1):55-65. doi: 10.1007/s10549-013-2667-y, PMID 23959397.
  71. Venuti V, Cannavà C, Cristiano MC, Fresta M, Majolino D, Paolino D, et al. A characterization study of resveratrol/sulfobutyl ether-b-cyclodextrin inclusion complex and in vitro anticancer activity. Colloids Surf B Biointerfaces. 2014;115:22-8. doi: 10.1016/j.colsurfb.2013.11.025, PMID 24321846.
  72. Neves AR, Queiroz JF, Reis S. Brain-targeted delivery of resveratrol using solid lipid nanoparticles functionalized with apolipoprotein E. J Nanobiotechnol. 2016;14(1):27. doi: 10.1186/s12951-016-0177-x.
  73. Khar Rk, Vyas SP 2002. Controlled Drug Delivery System-concepts and advances. 1st ed. New Delhi: Vallabh Prakashan.
  74. Kreuter JO. Historical Perspectives Nanoparticles-a historical perspective. Int J Pharm. 2007;331(1):1-10. doi: 10.1016/j.ijpharm.2006.10.021, PMID 17110063.
  75. Chrai SS, Murari R, Ahmed I. Liposomes, part II: drug delivery systems. Pharm Technol Eur. 2006:53-6.
  76. Daemen CD T, Scherphof GL. Vingerhoeds MH, Heeremans JLM; 1997. Liposomes: vesicles for the targeted and controlled delivery of peptides and proteins. Journal of Controlled Drug. Release 46. p. 165-75.
  77. Union E. Nanomedicines nanotechnology for Health European Technology Platform Strategic Reasearch Agenda for nanomedicines Union E, editor. November. European Union; 2006.

Reference

  1. Francisco JG, Ma LM, Paloma G, Ruth R. Nanotechnology and food industry. In:. In:Tech Dr. Valdez B, editor. Scientific, health and social aspects of the food industry. ISBN: 978-953-307-916-5; 2012. p. 1-35.
  2. Michele F, Oliveira, Pedro PG, Guimarães, Alinne DM, Gomes et al. Strategies to target tumors.
  3. Mohanraj VJ, Chen Y. Nanoparticles - a review. Trop J Pharm Res. 2006;5(1):561-73. doi: 10.4314/tjpr.v5i1.14634.
  4. Hoet PHM, Hohlfeld IB, Salata OV. Nanoparticles -known and unknown health risks. J Nano Biotechnol. 2004;02:01-15.
  5. Thassu YPD. Drug delivery nanoparticles formulation and characterization. Drugs Pharm Sci. 2007;191.
  6. Adams M. The top ten technologies: where is nanotechnology? 2004;2004 [cited 2013 7/05/2013]; about scope & presence of Nanotechnology.
  7. Badjatya JK. Enhancement of Dissolution of Poorly water Soluble aceclofenac by preparing Nanoparticles Inventi Rapid. NDDS. 2012;2012:01-2.
  8. Mohmmed Ali DAAS, Showkat R, Wadood Siddiqui A. GPAT: A Ideal book for Pharmacy Competative Exams. 2nd ed GPAT: A Ideal; 2012.
  9. Moghimi SM, Kissel T. Particulate nanomedicines?. Adv Drug Deliv. 2006;58(14):1451-5. doi: 10.1016/j.addr.2006.09.010.
  10. Roger A. Nanotechnology applications for drug delivery. Drug Deliv Nanotechnol. 2004:21-8.
  11. des Rieux AD, Fievez V, Garinot M, Schneider YJ, Préat V. Nanoparticles as potential oral delivery systems of proteins and vaccines: A mechanistic approach. J Control Release. 2006;116(1):1-27. doi: 10.1016/j.jconrel.2006.08.013, PMID 17050027.
  12. Storm G, Crommelin DJA. Liposomes: quo vadis? Pharm Sci Technol Today. 1998;1(1):19-31. doi: 10.1016/S1461-5347(98)00007-8.
  13. Couvreur P, Vauthier C. Nanotechnology: intelligent design to treat complex diseases. Pharm Res. 2006;23(7):1417-50. doi: 10.1007/s11095-006-0284-8, PMID 16779701.
  14. Jain S, Shukla K, Jain V, Saraf S. Nanoparticles: emerging carriers for delivery of bioactive agents. Pharma Times; 2006. In: 39:30-5.
  15. Phillipos T. Nanoparticles safe! About .co [guide]; 2009.
  16. Michele F, Oliveira, Pedro PG, Guimarães, Alinne DM, Gomes et al. Strategies to target tumors.
  17. Rick D. Berkeley considering need for nano safety.articles.sfgate.com; 2006.
  18. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-74. doi: 10.1016/j.cell.2011.02.013, PMID 21376230.
  19. Baudino TA. Targeted cancer therapy: the next generation of cancer treatment. Curr Drug Discov Technol. 2015;12(1):3-20. doi: 10.2174/1570163812666150602144310, PMID 26033233.
  20. Shen B, Ma Y, Yu S, Ji C. Smart multifunctional magnetic nanoparticle-based drug delivery system for cancer thermo-chemotherapy and intracellular imaging. ACS Appl Mater Interfaces. 2016;8(37):24502-8. doi: 10.1021/acsami.6b09772, PMID 27573061.
  21. Huang B, Abraham WD, Zheng Y, Bustamante López SC, Luo SS, Irvine DJ. Active targeting of chemotherapy to disseminated tumors using nanoparticle-carrying T cells. Sci Transl Med. 2015;7(291):291ra94. doi: 10.1126/scitranslmed.aaa5447, PMID 26062846.
  22. Oerlemans C, Bult W, Bos M, Storm G, Nijsen JF, Hennink WE. Polymeric micelles in anticancer therapy: targeting, imaging and triggered release. Pharm Res. 2010;27(12):2569-89. doi: 10.1007/s11095-010-0233-4, PMID 20725771.
  23. Somani S, Dufès C. Applications of dendrimers for brain delivery and cancer therapy. Nanomedicine (Lond). 2014;9(15):2403-14. doi: 10.2217/nnm.14.130, PMID 25413857.
  24. Kaminskas LM, Boyd BJ, Porter CJ. Dendrimer pharmacokinetics: the effect of size, structure and surface characteristics on ADME properties. Nanomedicine (Lond). 2011;6(6):1063-84. doi: 10.2217/nnm.11.67, PMID 21955077.
  25. Quintana A, Raczka E, Piehler L, Lee I, Myc A, Majoros I, et al. Design and function of a dendrimerbased therapeutic nanodevice targeted to tumor cells through the folate receptor. Pharm Res. 2002;19(9):1310-6. doi: 10.1023/a:1020398624602, PMID 12403067.
  26. Baker Jr. JR. Dendrimer-based nanoparticles for cancer therapy. Hematology Am Soc Hematol Educ Program. 2009:708-19. doi: 10.1182/asheducation-2009.1.708, PMID 20008257.
  27. Cheng Y, Zhao L, Li Y, Xu T. Design of biocompatible dendrimers for cancer diagnosis and therapy: current status and future perspectives. Chem Soc Rev. 2011;40(5):2673-703. doi: 10.1039/c0cs00097c, PMID 21286593.
  28. Rastogi V, Yadav P, Bhattacharya SS, Mishra AK, Verma N, Verma A, et al. Carbon nanotubes: an emerging drug carrier for targeting cancer cells. J Drug Deliv. 2014;2014:670815. doi: 10.1155/2014/670815, PMID 24872894.
  29. Sanginario A, Miccoli B, Demarchi D. Carbon nanotubes as an effective opportunity for cancer diagnosis and treatment. Biosensors. 2017;7(1):9. doi: 10.3390/bios7010009, PMID 28212271.
  30. Dinesh B, Bianco A, Ménard-Moyon C. Designing multimodal carbon nanotubes by covalent multi-functionalization. Nanoscale. 2016;8(44):18596-611. doi: 10.1039/c6nr06728j, PMID 27805213.
  31. Murugesan S, Mousa SA, O’connor LJ, Lincoln DW, Linhardt RJ. Carbon inhibits vascular endothelial growth factor- and fibroblast growth factor-promoted angiogenesis. FEBS Lett. 2007;581(6):1157-60. doi: 10.1016/j.febslet.2007.02.022, PMID 17331505.
  32. Chen Z, Mao R, Liu Y. Fullerenes for cancer diagnosis and therapy: preparation, biological and clinical perspectives. Curr Drug Metab. 2012;13(8):1035-45. doi: 10.2174/138920012802850128, PMID 22380017.
  33. Prylutska SV, Skivka LM, Didenko GV, Prylutskyy YI, Evstigneev MP, Potebnya GP, et al. Complex of C60 fullerene with doxorubicin as a promising agent in antitumor therapy. Nanoscale Res Lett. 2015;10(1):499. doi: 10.1186/s11671-015-1206-7, PMID 26714861.
  34. Zhang Q, Yang W, Man N, Zheng F, Shen Y, Sun K, et al. Autophagy-mediated chemosensitization in cancer cells by fullerene C60 nanocrystal. Autophagy. 2009;5(8):1107-17. doi: 10.4161/auto.5.8.9842, PMID 19786831.
  35. Kolluru LP, Rizvi SAA, D’Souza M, D’Souza MJ. Formulation development of albumin based theragnostic nanoparticles as a potential tumor targeting and delivery system. J Drug Target. 2013;21(1):77-86. doi: 10.3109/1061186X.2012.729214, PMID 23036042.
  36. Wolfbeis OS. An overview of nanoparticles commonly used in fluorescent bioimaging. Chem Soc Rev. 2015;44(14):4743-68. doi: 10.1039/c4cs00392f, PMID 25620543.
  37. Emerich DF, Thanos CG. Targeted nanoparticle-based drug delivery and diagnosis. J Drug Target. 2007;15(3):163-83. doi: 10.1080/10611860701231810, PMID 17454354.
  38. Li J, Zhu JJ. Quantum dots for fluorescent biosensing and bio-imaging applications. Analyst. 2013;138(9):2506-15. doi: 10.1039/c3an36705c, PMID 23518695.
  39. Michalet X, Pinaud FF, Bentolila LA, Tsay JM, Doose S, Li JJ, et al. Quantum dots for live cells, in vivo imaging, and diagnostics. Science. 2005;307(5709):538-44. doi: 10.1126/science.1104274, PMID 15681376.
  40. Datta R, Jaitawat SS. Nanotechnology - the new frontier of medicine. Med J Armed Forces India. 2006;62(3):263-8. doi: 10.1016/S0377-1237(06)80016-X, PMID 27365690.
  41. Janib SM, Moses AS, MacKay JA. Imaging and drug delivery using theranostic nanoparticles. Adv Drug Deliv Rev. 2010;62(11):1052-63. doi: 10.1016/j.addr.2010.08.004, PMID 20709124.
  42. Bhojani MS, Van Dort M, Rehemtulla A, Ross BD. Targeted imaging and therapy of brain cancer using theranostic nanoparticles. Mol Pharm. 2010;7(6):1921-9. doi: 10.1021/mp100298r, PMID 20964352.
  43. Subrahmanyam CVS. Textbook of Physical Pharmaceutics (Thorouly revised & Enlarged). 2’nd Reprint (August) ed. Textbook of Physical Pharmaceutics. 1 Delhi: Vallabh Prakashan. 2005;388.
  44. Couvreur P, Vauthier C. Nanotechnology: intelligent design to treat complex diseases. Pharm Res. 2006;23(7):1417-50. doi: 10.1007/s11095-006-0284-8, PMID 16779701.
  45. Raval AJ, Madhabahi MP. Preparation and characterization of nanoparticles for solubility and dissolution rate enhancement of meloxicam. Int J Pharm. 2011;01:42-9.
  46. Moss JA. HIV/AIDS review. Radiol Technol. 2013;84(3):247-67; quiz p.268. PMID 23322863.
  47. Bartlett JG, Moore RD. Improving HIV therapy. Sci Am. 1998;279(1):84-7, 89. doi: 10.1038/scientificamerican0798-84, PMID 9648300.
  48. Mamo T, Moseman EA, Kolishetti N, Salvador-Morales C, Shi J, Kuritzkes DR, et al. Emerging nanotechnology approaches for HIV/AIDS treatment and prevention. Nanomedicine (Lond). 2010;5(2):269-85. doi: 10.2217/nnm.10.1, PMID 20148638.
  49. Khalil NM, Carraro E, Cótica LF, Mainardes RM. Potential of polymeric nanoparticles in AIDS treatment and prevention. Expert Opin Drug Deliv. 2011;8(1):95-112. doi: 10.1517/17425247.2011.543673, PMID 21143001.
  50. Crabtree-Ramírez B, Villasís-Keever A, Galindo-Fraga A, del Río C, Sierra-Madero J. Effectiveness of highly active antiretroviral therapy (HAART) among HIV-infected patients in Mexico. AIDS Res Hum Retroviruses. 2010;26(4):373-8. doi: 10.1089/aid.2009.0077, PMID 20377418.
  51. Shah LK, Amiji MM. Intracellular delivery of saquinavir in biodegradable polymeric nanoparticles for HIV/AIDS. Pharm Res. 2006;23(11):2638-45. doi: 10.1007/s11095-006-9101-7, PMID 16969696.
  52. Mallipeddi R, Rohan LC. Progress in antiretroviral drug delivery using nanotechnology. Int J Nanomedicine. 2010;5:533-47. PMID 20957115.
  53. Spudich SS, Ances BM. Central nervous system complications of HIV infection. Top Antivir Med. 2011;19(2):48-57. PMID 21868822.
  54. Rao KS, Ghorpade A, Labhasetwar V. Targeting anti-HIV drugs to the CNS. Expert Opin Drug Deliv. 2009;6(8):771-84. doi: 10.1517/17425240903081705, PMID 19566446.
  55. Nowacek AS, McMillan J, Miller R, Anderson A, Rabinow B, Gendelman HE. Nanoformulated antiretroviral drug combinations extend drug release and antiretroviral responses in HIV-1- infected macrophages: implications for neuroAIDS therapeutics. J Neuroimmune Pharmacol. 2010;5(4):592-601. doi: 10.1007/s11481-010-9198-7, PMID 20237859.
  56. Aggarwal BB, Van Kuiken ME, Iyer LH, Harikumar KB, Sung B. Molecular targets of nutraceuticals derived from dietary spices: potential role in suppression of inflammation and tumorigenesis. Exp Biol Med (Maywood). 2009;234(8):825-49. doi: 10.3181/0902-MR-78, PMID 19491364.
  57. Acosta E. Bioavailability of nanoparticles in nutrient and nutraceutical delivery. Curr Opin Colloid Interface Sci. 2009;14(1):3-15. doi: 10.1016/j.cocis.2008.01.002.
  58. McClements DJ, Li F, Xiao H. The nutraceutical bioavailability classification scheme: classifying nutraceuticals according to factors limiting their oral bioavailability. Annu Rev Food Sci Technol. 2015;6:299-327. doi: 10.1146/annurev-food-032814-014043, PMID 25705933.
  59. Mohanty C, Sahoo SK. The in vitro stability and in vivo pharmacokinetics of curcumin prepared as an aqueous nanoparticulate formulation. Biomaterials. 2010;31(25):6597-611. doi: 10.1016/j.biomaterials.2010.04.062, PMID 20553984.
  60. Carvalho DdM, Takeuchi KP, Geraldine RM, Moura CJd, Torres MCL. Production, solubility and antioxidant activity of curcumin nanosuspension. Food Sci Technol (Campinas). 2015;35(1):115-9. doi: 10.1590/1678-457X.6515.
  61. Shaikh J, Ankola DD, Beniwal V, Singh D, Kumar MN. Nanoparticle encapsulation improves oral bioavailability of curcumin by at least 9-fold when compared to curcumin administered with piperine as absorption enhancer. Eur J Pharm Sci. 2009;37(3-4):223-30. doi: 10.1016/j.ejps.2009.02.019, PMID 19491009.
  62. Sasaki H, Sunagawa Y, Takahashi K, Imaizumi A, Fukuda H, Hashimoto T, et al. Innovative preparation of curcumin for improved oral bioavailability. Biol Pharm Bull. 2011;34(5):660-5. doi: 10.1248/bpb.34.660, PMID 21532153.
  63. Celotti E, Ferrarini R, Zironi R, Conte LS. Resveratrol content of some wines obtained from dried Valpolicella grapes: recioto and Amarone. J Chromatogr A. 1996;730(1-2):47-52. doi: 10.1016/0021-9673(95)00962-0, PMID 8680595.
  64. Summerlin N, Soo E, Thakur S, Qu Z, Jambhrunkar S, Popat A. Resveratrol nanoformulations: challenges and opportunities. Int J Pharm. 2015;479(2):282-90. doi: 10.1016/j.ijpharm.2015.01.003, PMID 25572692.
  65. Walle T. Bioavailability of resveratrol. Ann N Y Acad Sci. 2011;1215:9-15. doi: 10.1111/j.1749-6632.2010.05842.x, PMID 21261636.
  66. Kapetanovic IM, Muzzio M, Huang Z, Thompson TN, McCormick DL. Pharmacokinetics, oral bioavailability, and metabolic profile of resveratrol and its dimethylether analog, pterostilbene, in rats. Cancer Chemother Pharmacol. 2011;68(3):593-601. doi: 10.1007/s00280-010-1525-4, PMID 21116625.
  67. Trela BC, Waterhouse AL. Resveratrol: isomeric molar absorptivities and stability. J Agric Food Chem. 1996;44(5):1253-7. doi: 10.1021/jf9504576.
  68. Sanna V, Siddiqui IA, Sechi M, Mukhtar H. Resveratrol-loaded nanoparticles based on poly(epsilon-caprolactone) and poly (D, L-lactic-co-glycolic acid)-poly (ethylene glycol) blend for prostate cancer treatment. Mol Pharm. 2013;10(10):3871-81. doi: 10.1021/mp400342f, PMID 23968375.
  69. Penalva R, Esparza I, Larraneta E, González-Navarro CJ, Gamazo C, Irache JM. Zein-based nanoparticles improve the oral bioavailability of resveratrol and its anti-inflammatory effects in a mouse model of endotoxic shock. J Agric Food Chem. 2015;63(23):5603-11. doi: 10.1021/jf505694e, PMID 26027429.
  70. Catania A, Barrajón-Catalán E, Nicolosi S, Cicirata F, Micol V. Immunoliposome encapsulation increases cytotoxic activity and selectivity of curcumin and resveratrol against HER2 overexpressing human breast cancer cells. Breast Cancer Res Treat. 2013;141(1):55-65. doi: 10.1007/s10549-013-2667-y, PMID 23959397.
  71. Venuti V, Cannavà C, Cristiano MC, Fresta M, Majolino D, Paolino D, et al. A characterization study of resveratrol/sulfobutyl ether-b-cyclodextrin inclusion complex and in vitro anticancer activity. Colloids Surf B Biointerfaces. 2014;115:22-8. doi: 10.1016/j.colsurfb.2013.11.025, PMID 24321846.
  72. Neves AR, Queiroz JF, Reis S. Brain-targeted delivery of resveratrol using solid lipid nanoparticles functionalized with apolipoprotein E. J Nanobiotechnol. 2016;14(1):27. doi: 10.1186/s12951-016-0177-x.
  73. Khar Rk, Vyas SP 2002. Controlled Drug Delivery System-concepts and advances. 1st ed. New Delhi: Vallabh Prakashan.
  74. Kreuter JO. Historical Perspectives Nanoparticles-a historical perspective. Int J Pharm. 2007;331(1):1-10. doi: 10.1016/j.ijpharm.2006.10.021, PMID 17110063.
  75. Chrai SS, Murari R, Ahmed I. Liposomes, part II: drug delivery systems. Pharm Technol Eur. 2006:53-6.
  76. Daemen CD T, Scherphof GL. Vingerhoeds MH, Heeremans JLM; 1997. Liposomes: vesicles for the targeted and controlled delivery of peptides and proteins. Journal of Controlled Drug. Release 46. p. 165-75.
  77. Union E. Nanomedicines nanotechnology for Health European Technology Platform Strategic Reasearch Agenda for nanomedicines Union E, editor. November. European Union; 2006.

Photo
Tushar Nikam
Corresponding author

Department of Pharmaceutics, Divine College of Pharmacy, Satana Nashik

Photo
Mansi Pawar
Co-author

Department of Pharmaceutics, Divine College of Pharmacy, Satana Nashik

Photo
Dhananjay Patil
Co-author

Department of Pharmaceutics, Divine College of Pharmacy, Satana Nashik

Photo
Yogesh Sharma
Co-author

Department of Pharmaceutics, Divine College of Pharmacy, Satana Nashik

Tushar Nikam, Mansi Pawar, Rutuja Nikam, Yogesh Sharma, Dhananjay Patil, Applications Of Nanoparticle System In Drug Delivery Technology, Int. J. of Pharm. Sci., 2024, Vol 2, Issue 5, 103-118. https://doi.org/10.5281/zenodo.11109591

More related articles
A Comprehensive Review On Analytical Methods For T...
Pallavi Turukmane, Charushila Bhangale, ...
A Pharmacological Evaluation Of Antihistaminic And...
Laxmi Sirvi, Rajendra S. Bapna, Ragini Bundela, Karunakar Shukla,...
A Review On- Artificial Intelligence In Pharmacy &...
Meet Shah, Swapnil G. Kale, Kalyani Raut, Sayali Gandhi, Swapnil ...
Research advance in clinical evaluation of antihypertensive Drug...
Vishavjeet Pisal, Tejaswini Kamble, Pritam Salokhe, Sachin Navale, Nilesh Chougule, ...
A Comprehensive Overview Of HPLC Method Development And Validation...
Sayali V. Ganjiwale, A. P. Dewani, A. V. Chandewar, ...
Advances Of NSAIDS In Ankylosing Spondylitis...
R. Meghana, A. V. Kishore Babu, A. Srinivasa Rao, ...
Related Articles
Formulation And Evaluation Of Herbal Anti-Bacterial Crack Heel Ointment Using Th...
Ms.Priyanka Raut, Mrs.Manisha Mishra, Gayatri Bawane, Ayushi Ambade, Alshapha Anjum Khan, Abhilasha ...
Pharmacovigilance In The Digital Age...
Shruti Aher, Manoj Mahajan, Aman Upaganlawar, Chandrashekhar Upasani, ...
Niosomes As A Potential Approach For Enhancing Topical Application...
Priyadarshini K, E Gopinath, Ganesh N. S., Vineeth Chandy, ...
Strategies And Challenges In Pharmaceutical Marketing...
Satyam Mahadev Pawar , Pritam Salokhe, Nilesh chougule , ...
More related articles
A Pharmacological Evaluation Of Antihistaminic And Anti Inflammatory Activity On...
Laxmi Sirvi, Rajendra S. Bapna, Ragini Bundela, Karunakar Shukla, ...
A Review On- Artificial Intelligence In Pharmacy & Applications Of Ai In Pharmac...
Meet Shah, Swapnil G. Kale, Kalyani Raut, Sayali Gandhi, Swapnil Katkhade, Rohini Haral, Sneha S. Ka...
A Pharmacological Evaluation Of Antihistaminic And Anti Inflammatory Activity On...
Laxmi Sirvi, Rajendra S. Bapna, Ragini Bundela, Karunakar Shukla, ...
A Review On- Artificial Intelligence In Pharmacy & Applications Of Ai In Pharmac...
Meet Shah, Swapnil G. Kale, Kalyani Raut, Sayali Gandhi, Swapnil Katkhade, Rohini Haral, Sneha S. Ka...