Next Article in Journal
A Combination Approach in Inhibiting Type 2 Diabetes-Related Enzymes Using Ecklonia radiata Fucoidan and Acarbose
Previous Article in Journal
Effect of UV Irradiation (A and C) on Casuarina equisetifolia-Mediated Biosynthesis and Characterization of Antimicrobial and Anticancer Activity of Biocompatible Zinc Oxide Nanoparticles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Promising Strategies of Colloidal Drug Delivery-Based Approaches in Psoriasis Management

1
Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
2
Department of Biochemistry, Primeasia University, 12- Kemal Ataturk Avenue, HBR Tower Banani C/A, Dhaka 1213, Bangladesh
3
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Shahbag, Dhaka 1000, Bangladesh
4
Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
5
Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
6
Deanship of Scientific Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
7
Department of Entomology, Rutgers University, New Brunswick, NJ 08901, USA
8
Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
9
Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
10
Department of Biochemistry and Molecular Biology, Trust University, Barishal, Ruiya, Nobogram Road, Barishal 8200, Bangladesh
*
Authors to whom correspondence should be addressed.
Pharmaceutics 2021, 13(11), 1978; https://doi.org/10.3390/pharmaceutics13111978
Submission received: 29 September 2021 / Revised: 28 October 2021 / Accepted: 9 November 2021 / Published: 22 November 2021

Abstract

:
Psoriasis is a chronic inflammatory autoimmune disorder that moderately affects social and interpersonal relationships. Conventional treatments for psoriasis have certain problems, such as poor drug penetration through the skin, hyper-pigmentation, and a burning sensation on normal and diseased skin. Colloidal drug delivery systems overcome the pitfalls of conventional approaches for psoriasis therapeutics and have improved patient safety parameters, compliance, and superior effectiveness. They also entail reduced toxicity. This comprehensive review’s topics include the pathogenesis of psoriasis, causes and types of psoriasis, conventional treatment alternatives for psoriasis, the need for colloidal drug delivery systems, and recent studies in colloidal drug delivery systems for the treatment of psoriasis. This review briefly describes colloidal drug delivery approaches, such as emulsion systems—i.e., multiple emulsion, microemulsion, and nano-emulsion; vesicular systems—i.e., liposomes, ethosomes, noisomes, and transferosomes; and particulate systems—i.e., solid lipid nanoparticles, solid lipid microparticles, nano-structured lipid carriers, dendrimers, nanocrystals, polymeric nanoparticles, and gold nanoparticles. The review was compiled through an extensive search of the literature through the PubMed, Google Scholar, and ScienceDirect databases. A survey of literature revealed seven formulations based upon emulsion systems, six vesicular drug delivery systems, and fourteen particulate systems reported for antipsoriatic drugs. Based on the literature studies of colloidal approaches for psoriasis management carried out in recent years, it has been concluded that colloidal pharmaceutical formulations could be investigated broadly and have a broad scope for effective management of many skin disorders in the coming decades.

1. Introduction

Psoriasis is a chronic inflammatory autoimmune disorder that moderately affects social and interpersonal relationships. It is rapidly built up by skin surface cells which quickly form itchy and painful red patches. An everlasting cure for psoriasis is not available; nevertheless, its effect can be decreased by quitting smoking, moisturizing, and stress management. Psoriasis is mainly proliferated by irregular keratinocyte differentiation and epidermal hyper-proliferation and is directly linked with diabetes and cardiovascular diseases [1,2,3]. It is an autoimmune acute or chronic disorder mediated by T-cells. It progresses by triggering the host’s immune system, which can be caused by keratin mediating cells multiplying and appearing on the skin’s surface. Usually, the keratinocyte cycle of development and replacement takes approximately 35–40 days, after which they shed off. However, in psoriasis, the maturation cycle takes about one week, and cells, instead of shedding off, accumulate on the skin’s surface to produce red lesions [4]. Biologics have been shown to be an excellent alternative therapy for people with moderate–severe psoriasis [5]. In the pathophysiology of psoriasis, tumor necrosis factor-α plays a crucial role. TNF-α levels are higher in psoriatics than in normal individuals, and the improvement is linked to the psoriasis area and severity index score. Psoriasis is increasingly regarded as a chronic inflammatory systemic illness mediated by multiple inflammatory cytokines, such as TNF-α, rather than a simple skin condition. TNF-α is key cytokine in psoriasis immune response activation, and also has important impacts on keratinocyte proliferation and the control of endothelium proteins required for T-cell migration [6]. Anti-TNF-α and anti-TNF-α receptor drugs are now being used to treat psoriasis [7].
Conventional treatments for psoriasis have certain problems, such as poor drug penetration through the skin, low aqueous solubility, poor bioavailability, asymmetric and erratic pharmacokinetic profiles, hyper-pigmentation, first-pass metabolism, and a burning sensation on normal and diseased skin. Colloidal drug delivery systems overcome the pitfalls of conventional approaches for psoriasis therapeutics. They have improved patient safety parameters, compliance, and superior effectiveness, in addition to reduced toxicity. In 2014, Marepally and colleagues incorporated two therapeutic nucleic acids, i.e., anti-STAT3 siRNA (siSTAT3) and anti-TNF-α siRNA (siTNF-α), into lipid nanoparticles using cationic amphiphilic oleyl chain-based lipids to develop fusogenic nucleic acid lipid particles (F-NALP). Topical delivery of F-NALP can transmit siSTAT3 and siTNF-α into the dermis and decrease the expression of STAT3 and TNF-α mRNAs to play synergistic role in treatment of psoriatic-like plaques [8]. Bessar et al., in 2016, developed methotrexate (MTX)-loaded gold nanoparticles functionalized with sodium 3-mercapto-1-propansulfonate (Au-3MPS@MTX conjugate). Compared to MTX alone, the conjugate was superiorly percutaneously absorbed following 24 h application on naked mouse skin, and therefore, could be explored for effective treatment of psoriasis [9]. Wan and co-researchers, in 2017, fabricated hybrid nanoparticles (FK506 NPs-NIC) composed of hyaluronic acid with cholesterol combined with nicotinamide for tacrolimus (FK506) and illustrated synergistic action on FK506 permeation through intact skin. The PASI score in a imiquimod-induced psoriasis model demonstrated that FK506 HA–Chol NP–NIC exerted an ameliorating effect on skin lesions superior to commercial FK506 ointment [10]. In 2018, Nemati et al. developed a non-toxic fusion peptide carrier, i.e., spherical nucleic acid gold nanoparticles (SNA-NCs) conjugate with siRNA, in order to enhance the penetration of siRNA into cells, and found that topical application of SNA-NCs siRNA improved psoriatic-like skin lesions via suppression of gene expression and T-cell production [11]. In 2018, a group of researchers investigated discoidal lipid nanoparticles of APTstat3 tagged with a 9-arginine cell-penetrating peptide (APTstat3-9R). APTstat3 is an inhibitor of signal transducer and activator of transcription-3 (STAT3). It was found that transcutaneous delivery of lipid nanoparticles accomplished proficient skin penetration and successfully reduced psoriatic skin inflammation without producing adverse systemic effects [12]. Ramalheiro et al., in 2020, generated encapsulated rapamycin using phytantriol-based cubosome-like liquid crystalline nanoparticles for transdermal and controlled release delivery, for psoriasis treatment, which showed a sustained drug release profile till 14 days and displayed in-vitro antiproliferative action in natural killer cells [13]. Recently, Fereig and colleagues, in 2014, developed chitosan nanoparticles with an anti-proliferative molecule, i.e., tacrolimus, which acts via T-lymphocytic cell inhibition. They reported skin deposition of 82% of the tacrolimus, which was significantly greater in comparison to pure Tacrolimus® ointment, which showed about 34.0% skin retention [14].
Jyothi et al. also described traditional treatments along with nano-carriers and herbals used in psoriasis [15]. The current article gives brief information about conventional treatments for psoriasis and updated comprehensive information on recent advancements in various types of nanotechnology based colloidal drug delivery systems for treatment of psoriasis. The current article encompasses the colloidal drug delivery systems in several categories—viz., emulsion systems, i.e., multiple emulsion, microemulsion, and nano-emulsion; vesicular systems, i.e., liposomes, ethosomes, noisomes, and transferosomes; particulate systems, i.e., solid lipid nanoparticles, solid lipid microparticles, nano-structured lipid carriers, dendrimers, nanocrystals, polymeric nanoparticles, and gold nanoparticles; and micelle systems, i.e., polymeric micelles, reversed micelles, and mixed micelles. Moreover, the recent advancements of such colloidal drug delivery systems, i.e., emulsion systems, vesicular systems, and particulate systems, have also been summarized in tabular form. Recent studies on herbal colloidal drug delivery systems for psoriasis management have also been incorporated in the present review. For this purpose, an extensive search of the literature was conducted using PubMed, Google Scholar, and ScienceDirect databases. The keywords used in the search strategy were “psoriasis”, “liposome”, “ethosome”, “niosome”, “transferosome”, “multiple emulsions”, “microemulsion”, “nano-emulsion”, “solid lipid nanoparticles”, “solid lipid microparticles”, “nanostructured lipid carrier”, “dendrimers”, “nanocrystals”, “polymeric nanoparticle” and “gold nanoparticle” in various combinations. This should help students and research scientists to better understand future research and development in the field of colloidal drug delivery-based treatment for psoriasis.

2. Pathogenesis of Psoriasis

It has been commonly recognized that dendritic cells (DCs) are competent antigen-presenting cells that undertake a substantial role in several preliminary phases of the disease. The triggering of DCs in psoriasis, nevertheless, is not essentially precise. Several suggested pathways comprise the identification of antimicrobial peptides (AMPs) secreted through keratinocytes in reaction to trauma, which typically become over-expressed within psoriatic skin. LL37, S100 proteins, and β-defensins are major psoriasis-associated AMPs that are liberated through injured keratinocytes. LL-37 forms a complex with self DNA and RNA to produce LL-37-DNA and LL37-RNA complexes. The LL-37-DNA complex stimulates plasmacytoid DC (pDC) via toll-like receptor (TLR)-9, which discharges interferon-α (IFN-α) and IFN-β, leading to myeloid DC (mDC) phenotype maturation followed by Th1 and Th17 differentiation and function. Th17 cells secrete interleukins (IL)-17, IL-21, and IL-22, which activate keratinocyte proliferation in the epidermis. Complex LL-37-RNA triggers pDC via the TLR-7 pathway, which undergoes the above-described phases. Activation of mDC occurs through the TLR-8 mechanism, which migrates into draining lymph nodes that secrete tumor necrosis factor (TNF)-α, IL-23, and IL-12. These cytokines activate Th1 and Th17 differentiation and functions. Th17 cells discharge IL-17, IL-21, and IL-22, stimulating keratinocyte proliferation in the epidermis (Figure 1) [16,17,18,19]. Genetic factors also play a primitive role in the pathogenesis of psoriasis. The genetic epidemiology mainly comprises factors such as twin studies, familial aggregation, heritability, susceptibility analysis, and pedigree analysis. The tendency of family aggregation in psoriasis is highly distinctive and varies according to the population. About 31.26% of the Chinese population has reported a family history of psoriasis—67% reported having primary relatives with the disease, and 47% having secondary relatives with it. Hence, genetic predisposition has been studied, and more than 80 susceptible loci have been identified to play roles in the pathogenesis of psoriasis. The related study of gene functioning is in full swing, and several modes of next generation sequencing technology are being used to develop more accurate and sensitive genetic markers which can be targeted by biologics to impart enhanced management of disease. The genetic findings have provided hints on the pathogenesis of the disease. Disease prevention and management have been focused on by developing advanced effective biologic treatments, in response. Some of the susceptible genes that can induce psoriasis are LCE cluster, AIM2, LRRC7, MTHFR, MGAT5, PSORS6, SLC12A8, PSORS5, PSORS 9, and LCE1-LCE6.

3. Causes and Types of Psoriasis

Stress and psoriasis are directly related to each other. Acute stress decreases one’s cortisol level, which mediates inflammation in the skin, leading to psoriasis expansion [20,21]. It has been found that psoriasis may be caused due to trauma-induced skin injuries and exposure to sunlight. Several factors, such as irritants, burns, skin tests, abrasions, and electrodesiccation, could enhance skin injuries [22,23,24,25,26]. This disease is characterized as a squamous cell disease epitomized by plaques, red, inflammatory, and silvery-white raised lesions one centimeter in diameter. The lesions on psoriasis patients that are present on the scalp are symmetrically spread. Psoriasis is normally present on the skin, scalp, elbows, sacral region, knees, tongue, or oral mucosa. On the tongue, yellow–white patches are present, which spread at a higher rate than most lesions, and they change daily. This condition is known as geographic tongue [27]. After puberty, an attack of microorganisms such as Malassezia furfur is responsible for seborrhea dermatitis on the scalp. The psoriatic lesions predispose all of the skin of the psoriatic patient to lesions. The part of the skin which does not involve lesions has idiopathic factors such as cell dilation and mononuclear cell infiltration [28]. Characteristic features of numerous psoriasis types, including plaques, guttate, pustular, erythrodermic, nail, and scalp psoriasis, are given in Table 1.

4. Conventional Treatment Alternatives for Psoriasis

Conventional psoriasis treatments consist of phototherapy, self-care, and medications that aim to confiscate psoriasis scales and prevent skin cells from growing promptly (Figure 2). Photodynamic therapy combines drugs with light therapy to destroy abnormal cells or close blood vessels. Self-care includes stress management; light therapy; ultraviolet light therapy; and applying petroleum jelly, coal tar extract, and moisturizer. Medications such as steroids work by reducing inflammation, slowing down the production of skin cells, and reducing itching. A vitamin A derivative unplugs blocked hair follicles and averts forming new blockages and decreases skin cell growth. Anti-inflammatory agents are mainly used to prevent or counteract inflammation in joints and tissues. Immunosuppressive drugs decrease the immune response. Vitamins helps with normal body functions, growth, and development. The current most effective topical treatment for psoriasis is a foam containing a steroid and a vitamin D derivative [35]. Nevertheless, all these conventional therapies have certain problems—i.e., poor drug penetration through the skin, hyper-pigmentation, and a burning sensation on normal and diseased skin in the case of topical therapy; and low aqueous solubility, poor bioavailability, and an asymmetric and erratic pharmacokinetic profile in the case of systemic therapy [1,2,3,15,26]. Systemic therapies based on traditional drugs such as cyclosporin and methotrexate, and biological therapies based on anti-tumor necrosis factor-alpha, anti-interleukin 17, and anti-interleukin 23 molecules, are used in severe cases with very good results [36,37].

5. The Need for Colloidal Drug Delivery Systems

Conventional treatments for psoriasis have certain problems, such as poor drug penetration through the skin, hyper-pigmentation, first-pass metabolism, and a burning sensation on normal and diseased skin. Therefore, colloidal drug delivery systems have been employed to overcome the pitfalls of conventional approaches for psoriasis treatment and improve patient safety parameters and effectiveness. Colloidal drug delivery systems have shown better drug diffusion and penetration, leading to enhanced and prolonged accumulation of drug within the skin. Moreover, drug targeting to epidermal and dermal sites can be attained, which leads to dose reduction. The colloidal formulations also minimize the systemic toxicity, burning, irritation, and necrotizing effects of certain drugs. Colloidal drug delivery has more tolerability and safety as compared to conventional treatments. The faster follicular and intercellular penetration pathway of the colloidal system is highly advantageous, as it avoids multiple administrations and improves therapeutic efficacy [1,4,15]. Figure 3 classifies various types of colloidal drug delivery systems that are highly effective for treating skin disorders. The comparative illustrations of characteristics of various colloidal carriers are depicted in Table 2 [38,39,40,41,42,43,44,45,46]. The benefits of colloidal carriers for topical drug delivery—which lead to improvements in therapeutic potential of medications for psoriasis treatment—have been summarized in Table 3 [47,48,49]. Table 4 summarizes the features of various carrier materials used in the production of colloidal systems.

6. Applications of Emulsion Drug Delivery Systems in Psoriasis

6.1. Multiple Emulsions

Emulsion drug delivery systems that can be applied for psoriasis treatments include multiple emulsion, microemulsion, and submicron emulsion systems (Figure 3). Recent advances in applications of emulsion drug delivery systems for psoriasis management are given in Table 5. Multiple emulsions are water-in-oil-in-water (w/o/w) or oil-in-water-in-oil (o/w/o) dispersed systems stabilized by hydrophilic or lipophilic surfactants. These emulsions demonstrate high potential for prolonged skin retention without enhancing its trans-dermal permeation, increased patient compliance, reduced side effects, and optimized therapeutic efficacy [38,39,40,41,42]. Laugel et al. prepared multiple topical emulsions of hydrocortisone, which showed prolonged drug release and 1.5 fold greater penetration of hydrocortisone through the epidermis compared to a simple emulsion [43,44].

6.2. Microemulsion

A microemulsion is a clear and stable system: an isotropic mixture of oil, water, and a co-surfactant with 10–100 nm globule diameters. Microemulsions are transparent or translucent, as their droplet diameter is less than a fourth of the wavelength of visible light. The microemulsion effect is governed by skin permeation, mobility, and release of drugs from the vehicle [62,63,64]. The permeation factor is influenced by alterations in the stratum cornea and numerous permeation enhancers, including short-chain fatty acids and isopropyl myristate [65]. Badawi et al. investigated salicylic microemulsions for enhanced solubility for productive topical keratolytic and antimicrobial activity [45]. Barolia et al. prepared a microemulsion of 8-methoxsalen and 5-methoxy psoralen for effective treatment of a hyper-proliferative skin disorder with improved accretion of the drug in the skin without side effects [46]. Behera et al. investigated a methoxsalen chitosan-coated microemulsion. They found decreased inflammation, prolonged drug release, and prolonged resistance of skin [59].

6.3. Nano-Emulsion

Submicron emulsions, also referred to as nano-emulsions and ultrafine emulsions, have droplets of 20–200 nm [66,67,68,69]. Drugs which have poor penetration and cause skin irritation can be delivered in nano-emulsion systems. Rajitha et al. prepared a chaulmoogra oil-loaded methotrexate nano-emulsion, which showed better skin diffusion, affluent skin retention, and 2-fold increases in antipsoriatic and anti-inflammatory properties as compared to methotrexate tablets or any marketed formulation [63]. Kaur et al. synthesized clobetasol propionate and a calcipotriol nano-emulsion to diminish inflammation as an antipsoriatic agent. They found prolonged release of the drug from the nano-emulsion gel compared to conventional medicine and any other marketed formulation [64].

7. Applications of Vesicular Drug Delivery Systems in Psoriasis

7.1. Liposomes

Vesicular drug delivery systems which can be beneficial for psoriasis treatments include liposomes, ethosomes, niosomes, pharmacosomes, phytosomes, and transferosomes (Figure 4). Current applications of the vesicular system for psoriasis management are given in Table 6. Liposomes are spherical-shaped small vesicles produced by cholesterol and natural, non-toxic phospholipids. These are amphiphilic molecules, as have both hydrophilic and hydrophobic ends. They are composed of bilayer components and provide rigidity and fluidity to liposome molecules (for example); unsaturated phosphatidylcholine provides permeability; and saturated phospholipids provide a rigid bilayer structure. A liposome can be a unilamellar vesicle—a single phospholipid bilayer sphere, or a multi-lamellar vesicle consisting of an onion structure [70]. Jain et al. prepared tacrolimus and curcumin-loaded synergistic liposphere gels, which showed enhanced suppression of interleukin-22, tumor necrosis factor-alpha, and interleukin-17, along with synergistic antipsoriatic activity [71]. Walunj et al. fabricated and investigated the superior efficacy of a topical gel comprising cyclosporine-loaded cationic liposomes in an imiquimod induced psoriatic plaque model [72].

7.2. Ethosomes and Niosomes

Ethanolic liposomes, referred to as ethosomes, are fabricated using ethanol as a solvent. Dubey et al. fabricated a methotrexate ethosomal gel which showed effectual antipsoriatic activity and good penetration through the skin [73]. Niosome vesicles are composed of a non-ionic surfactant. There are microscopic in size and have a lamellar structure. They can envelop a solute like liposomes, and have design flexibility. Niosomes are amphiphilic in nature, having both hydrophilic and hydrophobic characteristics. They protect the drug from the biological environment and ensure good availability (they allow little degradation). Niosomes can be used for trans-dermal and intracellular drug delivery, liver targeting, and psoriasis treatment [77,78,79]. Aggarwal et al. synthesized dithranol-loaded liposomes and niosomes, which provide localized drug delivery for psoriasis treatment along with dose reduction, thereby minimizing the burning, irritating, necrotizing, and staining effects of dithranol [75].

7.3. Transferosomes

Transferosomes are single-chain, surfactant-based, highly deface lipid vesicles that create an osmotic gradient driving force that transports the material through the skin. They provides quick invasion of the subcutaneous tissue via intracellular lipid pathway [80,81,82,83,84,85,86]. Lei et al. prepared a tacrolimus-loaded transferosome, which showed increased anti-atopic dermatitis activity compared to commercial tacrolimus ointment (Protopic®) liposomes-gel [75]. Gizaway et al. fabricated betamethasone di-propionate-loaded transferosomes that showed antipsoriatic activity, along with greater tolerability and better safety than the available tacrolimus formulation [76].

8. Applications of Particulate Drug Delivery Systems in Psoriasis

Particulate drug delivery systems include solid lipid nanoparticles (SLNs), solid lipid microparticles (SLMs), nanolipid carriers (NLCs), dendrimers, aquasomes, nanocrystals, polymeric nanoparticles, and gold nanoparticles (Figure 5). Current advancements of the particulate system for psoriasis therapy are described in Table 7.

8.1. Solid Lipid Nanoparticles (SLNs) and Solid Lipid Microparticles (SLMs)

SLNs and SLMs are colloidal, biodegradable, biocompatible, and water-based technology that avoids organic solvents. These are composed of lipids dispersed in aqueous solutions of surfactant and water. SLNs can control drug release, can improve drug stability, can confer drug targeting, have excellent biocompatibility, and can carry both lipophilic and hydrophilic drugs. SLNs are excellent topical agents because lipids are non-toxic and non-irritant, which can reduce inflammation and treat skin disorders such as psoriasis [101,102]. Ferreira et al. concluded that topical co-delivery of SLNs of methotrexate and etanercept could be a targeted approach for psoriasis management [87]. Pradhan et al. prepared fluocinolone acetonide-loaded solid lipid nanoparticles, and concluded that better penetration through the skin was achieved to treat psoriasis and hyper-proliferation keratinocytes. Furthermore, enhanced skin contact and ease of application were attained [88]. Abdel-Salam et al. prepared solid lipid nanoparticles of diflucortolone valerate and found increased anti-inflammation and skin retention compared to a normal gel [89].

8.2. Nano-Structured Lipid Carriers (NLCs)

NLCs enhance drug loading capacity and minimize drug expulsion to overcome the drawbacks of SLNs and SLMs. NLCs are composed of solid fatty acids and small amounts of liquid lipids [90,101,102,103,104]. Pradhan et al. fabricated fluocinolone acetonide loaded with nano-structured lipid carriers for effective and potential management of psoriasis [91]. Pinto et al. developed methotrexate-loaded nano-structured lipid carriers for achieving potential topical treatment of psoriasis with reduced systemic toxicity [92].

8.3. Dendrimers

Dendrimers are drug delivery systems consisting of highly branched molecules with a controlled, globular, reactive 3-dimensional structure with a large number of controlled peripheral functionalities. Drug molecules are either physically entrapped or covalently attached to functional groups to form drug–dendrimer conjugates [105]. Types of dendrimers include glycodendrimers, peptide dendrimers, and lysine core dendrimers. In glycodendrimers, a carbohydrate is incorporated into the dendrimer structure; i.e., the saccharine unit is present on the outer surface. In peptide dendrimers, lysine is incorporated in the core of the dendrimer structure [106]. Tripathi et al. prepared a dendrimer loaded with dithranol and found reduced burning sensation and skin irritation compared to conventional therapy [93]. Agrawal et al. explored the potential of dithranol-loaded polypropylene imine dendrimers, and found enhanced drug accumulation within the skin and enhanced targeting of epidermal and dermal sites for successful treatment of psoriasis [94,107].

8.4. Nanocrystals

Nanocrystals are nano-metric-sized crystals, i.e., 20–100 nm, which contain 100% pure drug without any conjugation with a polymer. Nanocrystals enhance solubility and dissolution by increasing the dissolution pressure, surface area, and curvature of particles, which are mainly responsible for improving the drug’s oral bioavailability [108]. Increased penetration is also achieved by nanocrystals due to the faster follicular and intercellular penetration pathway, which is highly advantageous. They have provided avoidance of multiple administration and increased residence time of the medicament [109]. Döge et al. prepared nanocrystal loaded with dexamethasone to treat lesions, and found that nanocrystal augmented the skin penetration of dexamethasone [96].

8.5. Polymeric and Gold Nanoparticles

Polymeric nanoparticles are colloidal carriers 10–1000 nm in diameter. The drug is either physically or chemically adsorbed on the surface or encapsulated inside the polymer. The silent characteristics of polymeric nanoparticles include a long shelf-life, small size, non-toxicity, and facilitating the therapeutic effect directly on the site of application. Polymeric nanoparticles can be synthesized from natural hydrophilic polymers, i.e., gelatin, albumin, alginate, chitosan, or a synthetic hydrophobic polymer—i.e., polystyrene or poly-methyl methacrylates [110]. Badilli et al. prepared poly-(d,l-lactic-co-glycolic acid) microspheres loaded with clobetasol propionate and formulated emugel, which has reduced side effects [96]. Anwer et al. developed poly (d,l-lactide-co-glycolide) nanoparticles loaded with apremilast, and found long-term retention suggesting a once-daily regimen [97]. Fratoddi et al. studied the in-vivo activity of methotrexate-loaded gold nanoparticles on an imiquimod-induced psoriasis-like mouse model, and found reductions in keratinocytes and epidermal thickness [98]. Madan et al. developed SLNs of mometasone furoate [99], and Agrawal et al. formulated NLCs of acitretin [100] to enhance the therapeutic effect of psoriasis management.

9. Applications of Micelle Drug Delivery Systems in Psoriasis

Micelle drug delivery systems include polymeric micelles, reverse micelles, and mixed micelles (Figure 6). Polymeric micelles are drug delivery systems with a diameter range of 10–100 nm consisting of hydrophobic cores and hydrophilic surfaces. Hydrophobic micelles are used for their stable structures, and their hydrophilic surfaces allow them to circulate in the blood for extended periods and remain unidentified by the reticuloendothelial system. Polymeric micelles can change the physicochemical structures and kinetics of drugs [111]. Polymeric micelle-incorporated drugs reach systemic circulation faster and have more excellent assembly in lymph nodes [112,113]. Lapteva et al. prepared tacrolimus-loaded polymeric micelles and concluded that tacrolimus micelles augment skin penetration by 2-fold as compared to a simple marketed formulation of tacrolimus [114].

10. Recent Advancements in Herbal Nano-Carriers for Psoriasis Treatment

The majority of herbal drugs are insoluble; hence, they have inadequate bioavailability and augmented systemic clearance, necessitating frequent administration or else high doses. Therefore, the development of a colloidal delivery system provides several benefits to phytoconstituents, such as improved permeability, reduced toxicity, augmented pharmacological strength, fortified stability, and sustained release. Consequently, the colloidal drug delivery systems of herbal ingredients have promise for improving the effects of, and conquering the troubles linked with, herbal medicines [115,116,117,118,119,120]. Meng et al. prepared a niosome gel loaded with celastrol and concluded that celastrol loaded into noisomes leads to a two-fold increase in drug penetration into the skin [119]. Pleguezuelos-Villa et al. developed a nano-emulsion loaded with magneferan to overcome the poor aqueous solubility and low bioavailability of magneferan, which is an anti-hyperproliferative and anti-inflammatory agent. It was concluded that the nano-emulsion improved bioavailability. Therefore, it could be a better treatment for inflammatory and skin disorders [121]. Divya et al. synthesized an acitretin and aloe-emodin-loaded chitin nanogel. They found deeper penetration of aloe-emodin into the tissue, thereby producing strong anti-inflammatory action to treat psoriasis [122]. Examples of several colloidal drug delivery systems for herbal constituents used for psoriasis management are represented in Table 8.

11. Conclusions

Conventional therapies for psoriasis treatment have specific problems—i.e., poor drug penetration through the skin, hyper-pigmentation, and a burning sensation on normal and diseased skin in cases of topical treatment; and low aqueous solubility, poor bioavailability, and asymmetric and erratic pharmacokinetic profiles in cases of systemic therapy. Colloidal drug delivery systems for available drugs provide better drug diffusion, penetration, and targeting, leading to greater and prolonged accumulation inside the skin. Besides, these colloidal formulations can minimize the doses; systemic toxicity; and burning, irritation, and necrotizing effects of medications available for skin disorders. In the coming years, research on colloidal drug delivery system-based drug products having effective drug carriers with targeted drug release features might be extensively explored to treat several skin disease conditions.

12. Current and Future Developments

Current treatments for psoriasis management have some issues, such as inadequate diffusion of the medication via the skin, hyper-pigmentation, and the feeling of burning on normal and infected tissue in cases of topical therapy; and low water solubility, inadequate bioavailability, and irregular pharmacokinetics in systemic therapies. However, colloidal systems have been developed for several drugs in recent years, as described in this review. Developing a colloidal drug delivery system for available drugs could improve drug delivery, diffusion, and targeting, which might contribute to accelerated and prolonged deposition within the skin. Furthermore, such colloidal compositions can reduce the dosages, and toxic effects, burning, itching, and necrosis, caused by existing medications. Investigations on colloidal drug carrier-based pharmaceutical formulations as efficient therapeutic agents with targeted drug release characteristics could be performed widely, and could have broad applicability for the effective management of many skin disorders in the coming decades.

Author Contributions

This work was carried out in collaboration with all authors. T.B. and M.S.R. share the correspondence duties. All authors have read and agreed to the published version of the manuscript.

Funding

The Deanship of Scientific Research (DSR) at King Abdulaziz University, Jeddah, Saudi Arabia has funded this project, under grant number FP-046-43.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors express gratitude to Chitkara College of Pharmacy, Chitkara University, Punjab, and India for compiling this review.

Conflicts of Interest

The authors declare no conflict of interest.

List of Abbreviations

DCDendritic cell
AMPAntimicrobial peptide
pDCPlasmacytoid dendritic cell
TLRToll-like receptor
mDCmyeloid dendritic cell
ILInterleukin
IFN-αInterferon-α
TPGSTocopherol polyethylene glycol 1000 succinate
DCDendritic cell
AMPAntimicrobial peptide
pDCPlasmacytoid dendritic cell
TLRToll-like receptor
mDCmyeloid dendritic cell
ILInterleukin
IFN-αInterferon-α
TPGSTocopherol polyethylene glycol 1000 succinate
siRNAsmall interfering RNAs

References

  1. Pradhan, M.; Alexander, A.; Singh, M.R.; Singh, D.; Saraf, S.; Saraf, S. Ajazuddin Understanding the prospective of nano-formulations towards the treatment of psoriasis. Biomed. Pharmacother. 2018, 107, 447–463. [Google Scholar] [CrossRef]
  2. Kim, W.B.; Jerome, D.; Yeung, J. Diagnostic et prise en charge du psoriasis. Can. Fam. Physician 2017, 63, e210. [Google Scholar]
  3. Raychaudhuri, S.K.; Maverakis, E.; Raychaudhuri, S.P. Diagnosis and classification of psoriasis. Autoimmun. Rev. 2014, 13, 490–495. [Google Scholar] [CrossRef]
  4. Sala, M.; Elaissari, A.; Fessi, H. Advances in psoriasis physiopathology and treatments: Up to date of mechanistic insights and perspectives of novel therapies based on innovative skin drug delivery systems (ISDDS). J. Control. Release 2016, 239, 182–202. [Google Scholar] [CrossRef] [PubMed]
  5. Rich, S.J.; Bello-Quintero, C.E. Advancements in the treatment of psoriasis: Role of biologic agents. J. Manag. Care Spec. Pharm. 2004, 10, 318–325. [Google Scholar] [CrossRef] [PubMed]
  6. Mehlis, S.L.; Gordon, K.B. The immunology of psoriasis and biologic immunotherapy. J. Am. Acad. Dermatol. 2003, 49, 44–50. [Google Scholar] [CrossRef]
  7. Takahashi, H.; Iizuka, H. Psoriasis and metabolic syndrome. J. Dermatol. 2012, 39, 212–218. [Google Scholar] [CrossRef] [Green Version]
  8. Marepally, S.; Boakye, C.H.; Patel, A.R.; Godugu, C.; Doddapaneni, R.; Desai, P.R.; Singh, M. Topical administration of dual siRNAs using fusogenic lipid nanoparticles for treating psoriatic-like plaques. Nanomedicine 2014, 9, 2157–2174. [Google Scholar] [CrossRef] [PubMed]
  9. Bessar, H.; Venditti, I.; Benassi, L.; Vaschieri, C.; Azzoni, P.; Pellacani, G.; Magnoni, C.; Botti, E.; Casagrande, V.; Federici, M.; et al. Functionalized gold nanoparticles for topical delivery of methotrexate for the possible treatment of psoriasis. Colloids Surf. B 2016, 141, 141–147. [Google Scholar] [CrossRef] [Green Version]
  10. Wan, T.; Pan, W.; Long, Y.; Yu, K.; Liu, S.; Ruan, W.; Pan, J.; Qin, M.; Wu, C.; Xu, Y. Effects of nanoparticles with hydrotropic nicotinamide on tacrolimus: Permeability through psoriatic skin and antipsoriatic and antiproliferative activities. Int. J. Nanomed. 2017, 12, 1485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Nemati, H.; Ghahramani, M.H.; Faridi-Majidi, R.; Izadi, B.; Bahrami, G.; Madani, S.H.; Tavoosidana, G. Using siRNA-based spherical nucleic acid nanoparticle conjugates for gene regulation in psoriasis. J. Control. Release 2017, 268, 259–268. [Google Scholar] [CrossRef] [PubMed]
  12. Kim, J.Y.; Ahn, J.; Kim, J.; Choi, M.; Jeon, H.; Choe, K.; Lee, D.Y.; Kim, P.; Jon, S. Nanoparticle-assisted transcutaneous delivery of a signal transducer and activator of transcription 3-inhibiting peptide ameliorates psoriasis-like skin inflammation. Am. Chem. Soc. Nano 2018, 12, 6904–6916. [Google Scholar] [CrossRef] [PubMed]
  13. Ramalheiro, A.; Paris, J.L.; Silva, B.F.; Pires, L.R. Rapidly dissolving microneedles for the delivery of cubosome-like liquid crystalline nanoparticles with sustained release of rapamycin. Int. J. Pharm. 2020, 591, 119942. [Google Scholar] [CrossRef] [PubMed]
  14. Fereig, S.A.; El-Zaafarany, G.M.; Arafa, M.G.; Abdel-Mottaleb, M.M. Tacrolimus-loaded chitosan nanoparticles for enhanced skin deposition and management of plaque psoriasis. Carbohydr. Polym. 2021, 268, 118238. [Google Scholar] [CrossRef]
  15. Jyothi, S.L.; Krishna, K.L.; Shirin, V.A.; Sankar, R.; Pramod, K.; Gangadharappa, H.V. Drug delivery systems for the treatment of psoriasis: Current status and prospects. J. Drug Deliv. Sci. Technol. 2021, 62, 102364. [Google Scholar] [CrossRef]
  16. Jordan, C.T.; Cao, L.; Roberson, E.D.O.; Duan, S.; Helms, C.A.; Nair, R.P.; Duffin, K.C.; Stuart, P.E.; Goldgar, D.; Hayashi, G.; et al. Rare and common variants in CARD14, encoding an epidermal regulator of NF-kappaB, in psoriasis. Am. J. Hum. Genet. 2012, 90, 796–808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Rendon, A.; Schäkel, K. Psoriasis pathogenesis and treatment. Int. J. Mol. Sci. 2019, 20, 1745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Nickoloff, B.J. Keratinocytes regain momentum as instigators of cutaneous inflammation. Trends Mol. Med. 2006, 12, 102–106. [Google Scholar] [CrossRef]
  19. Iizuka, H.; Honda, H.; Ishida-Yamamoto, A. Epidermal remodeling in psoriasis (II): A quantitative analysis of the epidermal architecture. J. Investig. Dermatol. 1997, 109, 806–810. [Google Scholar] [CrossRef] [Green Version]
  20. Alexopoulos, A.; Chrousos, G.P. Stress-related skin disorders. Rev. Endocr. Metab. Disord. 2016, 17, 295–304. [Google Scholar] [CrossRef]
  21. Schmid-Ott, G.; Jaeger, B.; Boehm, T.; Langer, K.; Stephan, M.; Raap, U.; Werfel, T. Immunological effects of stress in psoriasis. Br. J. Dermatol. 2009, 160, 782–785. [Google Scholar] [CrossRef] [PubMed]
  22. O’doherty, C.J.; Macintyre, C. Palmoplantar pustulosis and smoking. Br. Med. J. Clin. Res. Ed. 1985, 291, 861–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Tagami, H. Triggering factors. Clin. Dermatol. 1997, 15, 677–685. [Google Scholar] [CrossRef]
  24. Naldi, L.; Parazzini, P.; Brevi, A.; Peserico, A.; Veller Fornasa, C.; Grosso, G.; Rossi, E.; Marinaro, P.; Polenghi, M.M.; Finzi, A.; et al. Family history, smoking habits, alcohol consumption and risk of psoriasis. Br. J. Dermatol. 1992, 127, 212–217. [Google Scholar] [CrossRef] [PubMed]
  25. Powles, A.V.; Baker, B.S.; Rutman, A.J.; McFadden, J.P.; Valdimarsson, H.; Fry, L. Epidermal rupture is the initiating factor for the Koebner response in psoriasis. Acta Derm. Venereol. 1990, 70, 35–38. [Google Scholar]
  26. Fierlbeck, G.; Rassner, G.; Müller, C. Psoriasis Induced at the Injection Site of Recombinant Interferon Gamma: Results of Immunohistologic Investigations. Arch. Dermatol. 1990, 126, 351–355. [Google Scholar] [CrossRef]
  27. Langley, R.G.B.; Krueger, G.G.; Griffiths, C.E.M. Psoriasis: Epidemiology, clinical features, and quality of life. Ann. Rheum. Dis. 2005, 64, ii18–ii23. [Google Scholar] [CrossRef] [Green Version]
  28. Gaitanis, G.; Magiatis, P.; Hantschke, M.; Bassukas, I.D.; Velegraki, A. The Malassezia genus in skin and systemic diseases. Clin. Microbiol. Rev. 2012, 25, 106–141. [Google Scholar] [CrossRef] [Green Version]
  29. Krishnamurthy, K.; Walker, A.; Gropper, C.; Hoffman, C. To treat or not to treat? Management of guttate psoriasis and pityriasis rosea in patients with evidence of group A Streptococcal infection. J. Drugs Dermatol. 2010, 9, 241–250. [Google Scholar]
  30. Naldi, L.; Peli, L.; Parazzini, F.; Carrel, C.F. Family history of psoriasis, stressful life events, and recent infectious disease are risk factors for a first episode of acute guttate psoriasis: Results of a case-control study. J. Am. Acad. Dermatol. 2001, 44, 433–438. [Google Scholar] [CrossRef]
  31. Gooderham, M.J.; Van Voorhees, A.S.; Lebwohl, M.G. An update on generalized pustular psoriasis. Expert Rev. Clin. Immunol. 2019, 15, 907–919. [Google Scholar] [CrossRef] [Green Version]
  32. Erythrodermic (Exfoliative) Psoriasis. PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/8453895/ (accessed on 25 February 2021).
  33. Balasubramaniam, P.; Berth-Jones, J. Erythroderma: 90% skin failure. Hosp. Med. 2004, 65, 100–102. [Google Scholar] [CrossRef]
  34. Papp, K.; Berth-Jones, J.; Kragballe, K.; Wozel, G.; de la Brassinne, M. Scalp psoriasis: A review of current topical treatment options. J. Eur. Acad. Dermatol. Venereol. 2007, 21, 1151–1160. [Google Scholar] [CrossRef] [PubMed]
  35. Dattola, A.; Silvestri, M.; Bennardo, L.; Passante, M.; Rizzuto, F.; Dastoli, S.; Patruno, C.; Bianchi, L.; Nisticò, S.P. A novel vehicle for the treatment of psoriasis. Dermatol. Ther. 2020, 33, e13185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Amoruso, G.F.; Nisticò, S.P.; Iannone, L.; Russo, E.; Rago, G.; Patruno, C.; Bennardo, L. Ixekizumab may improve renal function in psoriasis. Healthcare 2021, 9, 543. [Google Scholar] [CrossRef] [PubMed]
  37. Dattola, A.; Silvestri, M.; Tamburi, F.; Amoruso, G.F.; Bennardo, L.; Nisticò, S.P. Emerging role of anti-IL23 in the treatment of psoriasis: When humanized is very promising. Dermatol. Ther. 2020, 33, e14504. [Google Scholar] [CrossRef]
  38. Lutz, R.; Aserin, A.; Wicker, L.; Garti, N. Release of electrolytes from W/O/W double emulsions stabilized by a soluble complex of modified pectin and whey protein isolate. Colloids Surf. B Biointerfaces 2009, 74, 178–185. [Google Scholar] [CrossRef] [PubMed]
  39. Nakashima, T.; Shimizu, M.; Kukizaki, M. Particle control of emulsion by membrane emulsification and its applications. Adv. Drug Deliv. Rev. 2000, 45, 47–56. [Google Scholar] [CrossRef]
  40. Márquez, A.L.; Medrano, A.; Panizzolo, L.A.; Wagner, J.R. Effect of calcium salts and surfactant concentration on the stability of water-in-oil (w/o) emulsions prepared with polyglycerol polyricinoleate. J. Colloid Interface Sci. 2010, 341, 101–108. [Google Scholar] [CrossRef]
  41. Bhatia, N.; Pandit, S.; Agrawal, S.; Gupta, D. A Review On Multiple Emulsions. Int. J. Pharm. erud. 2013, 3, 22–30. [Google Scholar]
  42. Prajapati, S.B.; Bhatt, H.; Koli, A.; Dharamsi, A.; Shah, S.A. An overview of Preparation, Evaluation and Applications of Multiple Emulsions. Int. J. Pharm. Res. Sch. 2013, 2, 142–150. [Google Scholar]
  43. Laugel, C.; Baillet, A.; Youenang Piemi, M.P.; Marty, J.P.; Ferrier, D. Oil-water-oil multiple emulsions for prolonged delivery of hydrocortisone after topical application: Comparison with simple emulsions. Int. J. Pharm. 1998, 160, 109–117. [Google Scholar] [CrossRef]
  44. Benigni, M.; Pescina, S.; Grimaudo, M.A.; Padula, C.; Santi, P.; Nicoli, S. Development of microemulsions of suitable viscosity for cyclosporine skin delivery. Int. J. Pharm. 2018, 545, 197–205. [Google Scholar] [CrossRef] [PubMed]
  45. Badawi, A.A.; Nour, S.A.; Sakran, W.S.; El-Mancy, S.M.S. Preparation and evaluation of microemulsion systems containing salicylic acid. AAPS PharmSciTech 2009, 10, 1081–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Baroli, B.; López-Quintela, M.A.; Delgado-Charro, M.B.; Fadda, A.M.; Blanco-Méndez, J. Microemulsions for topical delivery of 8-methoxsalen. J. Control. Release 2000, 69, 209–218. [Google Scholar] [CrossRef]
  47. Pradhan, M.; Singh, D.; Singh, M.R. Novel colloidal carriers for psoriasis: Current issues, mechanistic insight and novel delivery approaches. J. Control. Release 2013, 170, 380–395. [Google Scholar] [CrossRef] [PubMed]
  48. Kaul, S.; Gulati, N.; Verma, D.; Mukherjee, S.; Nagaich, U. Role of nanotechnology in cosmeceuticals: A review of recent advances. J. Pharm. 2018, 2018, 1–19. [Google Scholar] [CrossRef] [Green Version]
  49. Habib, S.; Singh, M. Recent Advances in Lipid-Based Nanosystems for Gemcitabine and Gemcitabine—Combination Therapy. Nanomaterials 2021, 11, 597. [Google Scholar] [CrossRef]
  50. Jaiswal, P.; Gidwani, B.; Vyas, A. Nanostructured lipid carriers and their current application in targeted drug delivery. Artif. Cells Nanomed. Biotechnol. 2016, 44, 27–40. [Google Scholar] [CrossRef]
  51. Algahtani, M.S.; Ahmad, M.Z.; Ahmad, J. Nanoemulsion loaded polymeric hydrogel for topical delivery of curcumin in psoriasis. J. Drug Deliv. Sci. Technol. 2020, 59, 101847. [Google Scholar] [CrossRef]
  52. Mittal, S.; Ali, J.; Baboota, S. Enhanced anti-psoriatic activity of tacrolimus loaded nanoemulsion gel via omega 3-Fatty acid (EPA and DHA) rich oils-fish oil and linseed oil. J. Drug Deliv. Sci. Technol. 2021, 63, 102458. [Google Scholar] [CrossRef]
  53. Gungor, S.; Rezigue, M. Nanocarriers mediated topical drug delivery for psoriasis treatment. Curr. Drug Metab. 2017, 18, 454–468. [Google Scholar] [CrossRef]
  54. Kapoor, D.N.; Bhatia, A.; Kaur, R.; Sharma, R.; Kaur, G.; Dhawan, S. PLGA: A unique polymer for drug delivery. Ther. Deliv. 2015, 6, 41–58. [Google Scholar] [CrossRef] [PubMed]
  55. Zhao, L.; Temelli, F.; Curtis, J.M.; Chen, L. Preparation of Liposomes Using Supercritical Carbon Dioxide Technology: Effects of Phospholipids and Sterols. Food Res. Int. 2015, 77, 63–72. [Google Scholar] [CrossRef]
  56. Ricci, M.; Oliva, R.; Del Vecchio, P.; Paolantoni, M.; Morresi, A.; Sassi, P. DMSO-induced Perturbation of Thermotropic Properties of Cholesterol-Containing DPPC Liposomes. Biochim. Biophys. Acta Biomembr. 2016, 1858, 3024–3031. [Google Scholar] [CrossRef] [PubMed]
  57. Saeed, R.M.; Dmour, I.; Taha, M.O. Stable chitosan-based nanoparticles using polyphosphoric acid or hexametaphosphate for tandem ionotropic/covalent crosslinking and subsequent investigation as novel vehicles for drug delivery. Front. Bioeng. Biotechnol. 2020, 8, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Aland, R.; Ganesan, M.; Rao, P.R.; Bhikshapathi, D.V. Solid Lipid Nanoparticles for Topical Delivery of Acitretin for the Treatment of Psoriasis by Design of Experiment. Int. J. Pharm. Sci. Nanotechnol. 2019, 12, 4474–4491. [Google Scholar] [CrossRef]
  59. Behera, J.; Behera, J.; Keservani, R.K.; Yadav, A.; Tripathi, M.; Chadoker, A. Methoxsalen loaded chitosan coated microemulsion for effective treatment of psoriasis. Int. J. Drug Deliv. 2011, 2, 159–167. [Google Scholar] [CrossRef] [Green Version]
  60. Rajitha, P.; Shammika, P.; Aiswarya, S.; Gopikrishnan, A.; Jayakumar, R.; Sabitha, M. Chaulmoogra oil based methotrexate loaded topical nanoemulsion for the treatment of psoriasis. J. Drug Deliv. Sci. Technol. 2019, 49, 463–476. [Google Scholar] [CrossRef]
  61. Kaur, A.; Katiyar, S.S.; Kushwah, V.; Jain, S. Nanoemulsion loaded gel for topical co-delivery of clobitasol propionate and calcipotriol in psoriasis. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 1473–1482. [Google Scholar] [CrossRef]
  62. Narang, A.S.; Delmarre, D.; Gao, D. Stable drug encapsulation in micelles and microemulsions. Int. J. Pharm. 2007, 345, 9–25. [Google Scholar] [CrossRef]
  63. Yuan, Y.; Li, S.-M.; Mo, F.-K.; Zhong, D.-F. Investigation of microemulsion system for transdermal delivery of meloxicam. Int. J. Pharm. 2006, 321, 117–123. [Google Scholar] [CrossRef]
  64. Olsson, U.; Lindman, B. Uni- and Bicontinuous Microemulsions. In The Structure, Dynamics and Equilibrium Properties of Colloidal Systems; Springer: Dordrecht, The Netherlands, 1990; pp. 233–242. [Google Scholar]
  65. Madhav, S.; Gupta, D. A review on microemulsion based system. Int. J. Pharm. Sci. Res. 2011, 2, 1888–1899. [Google Scholar]
  66. El-Aasser, M.S.; Sudol, E.D. Miniemulsions: Overview of research and applications. JCT Res. 2004, 1, 21–32. [Google Scholar]
  67. Tadros, T.; Izquierdo, P.; Esquena, J.; Solans, C. Formation and stability of nano-emulsions. Adv. Colloid Interface Sci. 2004, 108–109, 303–318. [Google Scholar] [CrossRef]
  68. Yu, H.; Huang, Q. Improving the oral bioavailability of curcumin using novel organogel-based nanoemulsions. J. Agric. Food Chem. 2012, 60, 5373–5379. [Google Scholar] [CrossRef]
  69. Singh, Y.; Meher, J.G.; Raval, K.; Khan, F.A.; Chaurasia, M.; Jain, N.K.; Chourasia, M.K. Nanoemulsion: Concepts, development and applications in drug delivery. J. Control. Release 2017, 252, 28–49. [Google Scholar] [CrossRef] [PubMed]
  70. Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S.W.; Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K. Liposome: Classification, preparation, and applications. Nanoscale Res. Lett. 2013, 8, 102. [Google Scholar] [CrossRef] [Green Version]
  71. Jain, A.; Doppalapudi, S.; Domb, A.J.; Khan, W. Tacrolimus and curcumin co-loaded liposphere gel: Synergistic combination towards management of psoriasis. J. Control. Release 2016, 243, 132–145. [Google Scholar] [CrossRef]
  72. Walunj, M.; Doppalapudi, S.; Bulbake, U.; Khan, W. Preparation, characterization, and in vivo evaluation of cyclosporine cationic liposomes for the treatment of psoriasis. J. Liposome Res. 2020, 30, 68–79. [Google Scholar] [CrossRef]
  73. Dubey, V.; Mishra, D.; Dutta, T.; Nahar, M.; Saraf, D.K.; Jain, N.K. Dermal and transdermal delivery of an anti-psoriatic agent via ethanolic liposomes. J. Control. Release 2007, 123, 148–154. [Google Scholar] [CrossRef] [PubMed]
  74. Agarwal, R.; Katare, O.P.; Vyas, S.P. Preparation and in vitro evaluation of liposomal/niosomal delivery systems for antipsoriatic drug dithranol. Int. J. Pharm. 2001, 228, 43–52. [Google Scholar] [CrossRef]
  75. Lei, W.; Yu, C.; Lin, H.; Zhou, X. Development of tacrolimus-loaded transfersomes for deeper skin penetration enhancement and therapeutic effect improvement in vivo. Asian J. Pharm. Sci. 2013, 8, 336–345. [Google Scholar] [CrossRef] [Green Version]
  76. EI Gizaway, S.; Fadel, M.; Mourad, B.; Elnaby, F.E.A. Betamethasone dipropionate gel for treatment of localized plaque psoriasis. Int. J. Pharm. Pharm. Sci. 2017, 9, 173. [Google Scholar] [CrossRef] [Green Version]
  77. Radha, G.; Rani, T.S.; Sarvani, B. A review on proniosomal drug delivery system for targeted drug action. J. Basic Clin. Pharm. 2013, 4, 42. [Google Scholar] [CrossRef] [Green Version]
  78. Khan, R.; Irchhaiya, R. Niosomes: A potential tool for novel drug delivery. J. Pharm. Investig. 2016, 46, 195–204. [Google Scholar] [CrossRef]
  79. Lakshmi, P.K.; Devi, G.S.; Bhaskaran, S.; Sacchidanand, S. Niosomal methotrexate gel in the treatment of localized psoriasis: Phase I and phase II studies. Indian J. Dermatol. Venereol. Leprol. 2007, 73, 157–161. [Google Scholar] [CrossRef]
  80. Semalty, A.; Semalty, M.; Rawat, B.S.; Singh, D.; Rawat, M.S.M. Pharmacosomes: The lipid-based new drug delivery system. Expert Opin. Drug Deliv. 2009, 6, 599–612. [Google Scholar] [CrossRef]
  81. Meshram, S.S.; Itankar, P.R.; Patil, A.T.; Meshram, M.S.S. To Study Pharmacognostic, Physicochemical and Phytochemical Study of Stem Bark of Bauhinia purpurea Linn. J. Pharmacogn. Phytochem. 2013, 2, 19–22. [Google Scholar]
  82. Jain, N.; Gupta, B.P.; Thakur, N.; Jain, R.; Banweer, J.; Jain, D.K.; Jain, S. Phytosome: A Novel Drug Delivery System for Herbal Medicine. Int. J. Pharm. Sci. Drug Res. 2010, 2, 224–228. [Google Scholar]
  83. Cevc, G.; Schätzlein, A.; Blume, G. Transdermal drug carriers: Basic properties, optimization and transfer efficiency in the case of epicutaneously applied peptides. J. Control. Release 1995, 36, 3–16. [Google Scholar] [CrossRef]
  84. Lin, H.W.; Xie, Q.C.; Huang, X.; Ban, J.F.; Wang, B.; Wei, X.; Chen, Y.Z.; Lu, Z.F. Increased skin permeation efficiency of imperatorin via charged ultradeformable lipid vesicles for transdermal delivery. Int. J. Nanomed. 2018, 13, 831–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Rai, S.; Pandey, V.; Rai, G. Transfersomes as versatile and flexible nano-vesicular carriers in skin cancer therapy: The state of the art. Nano Rev. Exp. 2017, 8, 1325708. [Google Scholar] [CrossRef] [PubMed]
  86. Ferreira, M.; Barreiros, L.; Segundo, M.A.; Torres, T.; Selores, M.; Costa Lima, S.A.; Reis, S. Topical co-delivery of methotrexate and etanercept using lipid nanoparticles: A targeted approach for psoriasis management. Colloids Surf. B Biointerfaces 2017, 159, 23–29. [Google Scholar] [CrossRef]
  87. Pradhan, M.; Singh, D.; Singh, M.R. Development characterization and skin permeating potential of lipid based novel delivery system for topical treatment of psoriasis. Chem. Phys. Lipids 2015, 186, 9–16. [Google Scholar] [CrossRef]
  88. Abdel-Salam, F.S.; Elkheshen, S.A.; Mahmoud, A.A.; Ammar, H.O. Diflucortolone valerate loaded solid lipid nanoparticles as a semisolid topical delivery system. Bull. Fac. Pharm. Cairo Univ. 2016, 54, 1–7. [Google Scholar] [CrossRef] [Green Version]
  89. Arora, R.; Katiyar, S.S.; Kushwah, V.; Jain, S. Solid lipid nanoparticles and nanostructured lipid carrier-based nanotherapeutics in treatment of psoriasis: A comparative study. Expert Opin. Drug Deliv. 2017, 14, 165–177. [Google Scholar] [CrossRef] [PubMed]
  90. Pradhan, M.; Singh, D.; Murthy, S.N.; Singh, M.R. Design, characterization and skin permeating potential of Fluocinolone acetonide loaded nanostructured lipid carriers for topical treatment of psoriasis. Steroids 2015, 101, 56–63. [Google Scholar] [CrossRef]
  91. Pinto, M.F.; Moura, C.C.; Nunes, C.; Segundo, M.A.; Costa Lima, S.A.; Reis, S. A new topical formulation for psoriasis: Development of methotrexate-loaded nanostructured lipid carriers. Int. J. Pharm. 2014, 477, 519–526. [Google Scholar] [CrossRef]
  92. Tripathi, P.K.; Gorain, B.; Choudhury, H.; Srivastava, A.; Kesharwani, P. Dendrimer entrapped microsponge gel of dithranol for effective topical treatment. Heliyon 2019, 5, e01343. [Google Scholar] [CrossRef] [Green Version]
  93. Agrawal, U.; Mehra, N.K.; Gupta, U.; Jain, N.K. Hyperbranched dendritic nano-carriers for topical delivery of dithranol. J. Drug Target. 2013, 21, 497–506. [Google Scholar] [CrossRef]
  94. Döge, N.; Hönzke, S.; Schumacher, F.; Balzus, B.; Colombo, M.; Hadam, S.; Rancan, F.; Blume-Peytavi, U.; Schäfer-Korting, M.; Schindler, A.; et al. Ethyl cellulose nanocarriers and nanocrystals differentially deliver dexamethasone into intact, tape-stripped or sodium lauryl sulfate-exposed ex vivo human skin—Assessment by intradermal microdialysis and extraction from the different skin layers. J. Control. Release 2016, 242, 25–34. [Google Scholar] [CrossRef] [PubMed]
  95. Badıllı, U.; Şen, T.; Tarımcı, N. Microparticulate based topical delivery system of clobetasol propionate. AAPS PharmSciTech 2011, 12, 949–957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Anwer, M.K.; Mohammad, M.; Ezzeldin, E.; Fatima, F.; Alalaiwe, A.; Iqbal, M. Preparation of sustained release apremilast-loaded PLGAlga nanoparticles: In vitro characterization and in vivo pharmacokinetic study in rats. Int. J. Nanomed. 2019, 14, 1587–1595. [Google Scholar] [CrossRef] [Green Version]
  97. Fratoddi, I.; Benassi, L.; Botti, E.; Vaschieri, C.; Venditti, I.; Bessar, H.; Samir, M.A.; Azzoni, P.; Magnoni, C.; Costanzo, A.; et al. Effects of topical methotrexate loaded gold nanoparticle in cutaneous inflammatory mouse model. Nanomed. Nanotechnol. Biol. Med. 2019, 17, 276–286. [Google Scholar] [CrossRef] [PubMed]
  98. Madan, J.; Dua, K.; Khude, P. Development and evaluation of solid lipid nanoparticles of mometasone furoate for topical delivery. Int. J. Pharm. Investig. 2014, 4, 60. [Google Scholar] [CrossRef] [Green Version]
  99. Agrawal, Y.; Petkar, K.C.; Sawant, K.K. Development, evaluation and clinical studies of Acitretin loaded nanostructured lipid carriers for topical treatment of psoriasis. Int. J. Pharm. 2010, 401, 93–102. [Google Scholar] [CrossRef] [PubMed]
  100. Mukherjee, S.; Ray, S.; Thakur, R.S. Solid lipid nanoparticles: A modern formulation approach in drug delivery system. Indian J. Pharm. Sci. 2009, 71, 349–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Müller, R.H.; Mäder, K.; Gohla, S. Solid lipid nanoparticles (SLN) for controlled drug delivery—A review of the state of the art. Eur. J. Pharm. Biopharm. 2000, 50, 161–177. [Google Scholar] [CrossRef]
  102. Umeyor, E.C.; Kenechukwu, F.; Ogbonna, J.D.; Chime, S.; Attama, A. Investigation of solidified reverse micellar systems as novel carriers for oral delivery of gentamicin. J. Pharm. Res. 2012, 5, 4914–4920. [Google Scholar]
  103. Gaba, B.; Fazil, M.; Ali, A.; Baboota, S.; Sahni, J.K.; Ali, J. Nanostructured lipid (NLCs) carriers as a bioavailability enhancement tool for oral administration. Drug Deliv. 2015, 22, 691–700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Müller, R.H.; Radtke, M.; Wissing, S.A. Nanostructured lipid matrices for improved microencapsulation of drugs. Int. J. Pharm. 2002, 242, 121–128. [Google Scholar] [CrossRef]
  105. Liu, M.; Fréchet, J.M.J. Designing dendrimers for drug delivery. Pharm. Sci. Technol. Today 1999, 2, 393–401. [Google Scholar] [CrossRef]
  106. Cloninger, M.J. Biological applications of dendrimers. Curr. Opin. Chem. Biol. 2002, 6, 742–748. [Google Scholar] [CrossRef]
  107. Narang, N. Aquasomes: Self-assembled systems for the delivery of bioactive molecules. Asian J. Pharm. 2014, 6, 95. [Google Scholar] [CrossRef]
  108. Müller, R. Junghanns Nanocrystal technology, drug delivery and clinical applications. Int. J. Nanomed. 2008, 3, 295. [Google Scholar] [CrossRef] [Green Version]
  109. Raj, S.; Jose, S.; Sumod, U.S.; Sabitha, M. Nanotechnology in cosmetics: Opportunities and challenges. J. Pharm. Bioallied Sci. 2012, 4, 186–193. [Google Scholar] [CrossRef]
  110. Safari, J.; Zarnegar, Z. Advanced drug delivery systems: Nanotechnology of health design A review. J. Saudi Chem. Soc. 2014, 18, 85–99. [Google Scholar] [CrossRef]
  111. Yadav, H.K. Different techniques for preparation of polymeric nanoparticles—A review. Asian J. Pharm. Clin. Res. 2012, 5, 16–23. [Google Scholar]
  112. Xu, W.; Ling, P.; Zhang, T. Polymeric Micelles, a Promising Drug Delivery System to Enhance Bioavailability of Poorly Water-Soluble Drugs. J. Drug Deliv. 2013, 2013, 1–15. [Google Scholar] [CrossRef]
  113. Ahmad, Z.; Shah, A.; Siddiq, M.; Kraatz, H.B. Polymeric micelles as drug delivery vehicles. RSC Adv. 2014, 4, 17028–17038. [Google Scholar] [CrossRef]
  114. Gaucher, G.; Dufresne, M.H.; Sant, V.P.; Kang, N.; Maysinger, D.; Leroux, J.C. Block copolymer micelles: Preparation, characterization and application in drug delivery. J. Control. Release 2005, 109, 169–188. [Google Scholar] [CrossRef] [PubMed]
  115. Lapteva, M.; Mondon, K.; Möller, M.; Gurny, R.; Kalia, Y.N. Polymeric micelle nanocarriers for the cutaneous delivery of tacrolimus: A targeted approach for the treatment of psoriasis. Mol. Pharm. 2014, 11, 2989–3001. [Google Scholar] [CrossRef] [PubMed]
  116. Novel Approach: Herbal Remedies and Natural Products. Google Scholar. Available online: https://scholar.google.com/scholar?hl=en&as_sdt=0%2C5&q=Novel+approach%3A+Herbal+remedies+and+natural+products+in+pharmaceutical+science+as+nano+drug+delivery+systems.+&btnG= (accessed on 26 February 2021).
  117. Shirwaikar, A.; Shirwaikar, A.; Prabhu, S.; Kumar, G. Herbal excipients in novel drug delivery systems. Indian J. Pharm. Sci. 2008, 70, 415–422. [Google Scholar] [CrossRef] [Green Version]
  118. Sungthongjeen, S.; Pitaksuteepong, T.; Somsiri, A.; Sriamornsak, P. Studies on pectins as potential hydrogel matrices for controlled-release drug delivery. Drug Dev. Ind. Pharm. 1999, 25, 1271–1276. [Google Scholar] [CrossRef] [PubMed]
  119. Promising Role of Nanopharmaceuticals in Drug Delivery. Google Scholar. Available online: https://scholar.google.com/scholar?hl=en&as_sdt=0%2C5&q=Promising+role+of+nanopharmaceuticals+in+drug+delivery&btnG= (accessed on 26 February 2021).
  120. Li, Y.; Dong, L.; Jia, A.; Chang, X.; Xue, H. Preparation and characterization of solid lipid nanoparticles loaded traditional chinese medicine. Int. J. Biol. Macromol. 2006, 38, 296–299. [Google Scholar] [CrossRef]
  121. Fasinu, P.; Pillay, V.; Ndesendo, V.M.K.; Du Toit, L.C.; Choonara, Y.E. Diverse approaches for the enhancement of oral drug bioavailability. Biopharm. Drug Dispos. 2011, 32, 185–209. [Google Scholar] [CrossRef]
  122. Mainardi, T.; Kapoor, S.; Bielory, L. Complementary and alternative medicine: Herbs, phytochemicals and vitamins and their immunologic effects. J. Allergy Clin. Immunol. 2009, 123, 283–294. [Google Scholar] [CrossRef]
  123. Meng, S.; Sun, L.; Wang, L.; Lin, Z.; Liu, Z.; Xi, L.; Wang, Z.; Zheng, Y. Loading of water-insoluble celastrol into niosome hydrogels for improved topical permeation and anti-psoriasis activity. Colloids Surf. B Biointerfaces 2019, 182, 1–9. [Google Scholar] [CrossRef]
  124. Pleguezuelos-Villa, M.; Nácher, A.; Hernández, M.J.; Ofelia Vila Buso, M.A.; Ruiz Sauri, A.; Díez-Sales, O. Mangiferin nanoemulsions in treatment of inflammatory disorders and skin regeneration. Int. J. Pharm. 2019, 564, 299–307. [Google Scholar] [CrossRef]
  125. Divya, G.; Panonnummal, R.; Gupta, S.; Jayakumar, R.; Sabitha, M. Acitretin and aloe-emodin loaded chitin nanogel for the treatment of psoriasis. Eur. J. Pharm. Biopharm. 2016, 107, 97–109. [Google Scholar] [CrossRef] [PubMed]
  126. Sonia, K.; Anupama, D. Microemulsion Based Transdermal Drug Delivery of Tea Tree Oil. Int. J. Drug Dev. Res. 2010, 3, 191–198. [Google Scholar]
  127. Filippone, A.; Consoli, G.M.L.; Granata, G.; Casili, G.; Lanza, M.; Ardizzone, A.; Cuzzocrea, S.; Esposito, E.; Paterniti, I. Topical delivery of curcumin by choline-calix[4]arene-based nanohydrogel improves its therapeutic effect on a psoriasis mouse model. Int. J. Mol. Sci. 2020, 21, 5053. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathogenesis of psoriasis.
Figure 1. Pathogenesis of psoriasis.
Pharmaceutics 13 01978 g001
Figure 2. Conventional treatment alternatives for psoriasis treatment.
Figure 2. Conventional treatment alternatives for psoriasis treatment.
Pharmaceutics 13 01978 g002
Figure 3. Types of colloidal drug delivery systems.
Figure 3. Types of colloidal drug delivery systems.
Pharmaceutics 13 01978 g003
Figure 4. Basic structures of emulsion and vesicular drug delivery systems.
Figure 4. Basic structures of emulsion and vesicular drug delivery systems.
Pharmaceutics 13 01978 g004
Figure 5. Basic structures of particulate drug delivery systems.
Figure 5. Basic structures of particulate drug delivery systems.
Pharmaceutics 13 01978 g005
Figure 6. Basic structures of micelle drug delivery systems.
Figure 6. Basic structures of micelle drug delivery systems.
Pharmaceutics 13 01978 g006
Table 1. Types of psoriasis and their characteristic features.
Table 1. Types of psoriasis and their characteristic features.
Psoriasis Type
(Affected Area)
Characteristic FeaturesRef.
Guttate psoriasis (Head and limbs)The lesions are monomorphic. Children and adolescents are predominantly affected. The upper respiratory tract infection is occurring followed by streptococcal infection. [15,29,30]
Plaques psoriasis/Psoriasis vulgaris (all body but typically on elbows, knees, scalp areas)The characteristic lesions are dry, sharp and oval in shape which seems as erythematous macules and form plaque. [15]
Pustular psoriasis
(Palms and soles)
The patient suffers from red, inflamed, skin. In localized pustulosis, two distinct types of psoriasis are acrodermatitis continua of hallopeau and palmoplantar pustulosis in which pustules are formed which spread all over feet. In generalized psoriasis, pustular lesions occur in pregnancy state or due to some drugs. [31]
Erythrodermic psoriasis
(All body)
This is unstable psoriasis which is occurred by various reasons like cardiac failure, hyperthermia or deficiency of vitamins [32,33]
Nail psoriasis
(Fingernail, toe-nails)
At proximal portion of nail, small pits are forms which are characterized by orange-yellow area below nail plate which is called as oil spots. [3]
Scalp psoriasis
(Hairline)
Selection of appropriate treatment is difficult to need of application at scalp area. [34]
Table 2. Distinguishing features among different types of colloidal nano-carriers.
Table 2. Distinguishing features among different types of colloidal nano-carriers.
ParameterPhytosomeLiposomeSolid Lipid NanoparticlesPolymeric
Nanoparticle
BondPresentAbsentAbsentAbsent
Complexity in manufacturingLess complexMore ComplexMore ComplexMore Complex
Drug leakageLessMoreMoreMore
Lipid drug interactionYesNoNoNo
StabilityMore stableLess StableLess StableMore Stable
EntrapmentHighLowLowLow
Nature of excipientsLipidLipidLipidSynthetic or natural polymer
BiocompatibilityHighHighHighLow
Safety profile of solventsICH Class III solventsICH Class II and III solventsICH Class II and III solventsICH Class II and III solvents
BioavailabilityHighModerateModerateModerate
AbsorptionHighModerateModerateModerate
Table 3. Advantages of colloidal drug delivery systems for topical drug delivery for psoriasis treatment.
Table 3. Advantages of colloidal drug delivery systems for topical drug delivery for psoriasis treatment.
Nanocarrier SystemAdvantages
Vesicular drug delivery system
TransferosomeLarge molecular weight medications are delivered to skin in a non-invasive manner.
EthosomeHighly permeable and compliance to patient as well as safer for skin
LiposomeAmphiphilic nature, biocompatibility, and ease of surface alteration
PhytosomesGreat oral and transdermal bioavailability and therapeutic effect
NiosomeEnhances skin penetration of drugs; improved bioavailability of insufficiently absorbed drugs
Particulate drug delivery system
Solid lipid nanoparticleIncreased efficacy, biocompatible, biodegradable
DendrimersEasy to prepare and alterations; better skin penetration
Polymeric nanoparticle Used for entrapment of various class of drugs; biocompatible, biodegradable
Nanostructured lipid carriersReduces drug leakage during storage, increases drug payload; biocompatible
Micelles drug delivery systems
Polymeric micelles Thermodynamic stability, self-assembling, and skin targeting potential.
Table 4. Characteristics of carriers/matrices utilized in the production of colloidal drug delivery systems.
Table 4. Characteristics of carriers/matrices utilized in the production of colloidal drug delivery systems.
Carrier/Matrix for NanocarrierPropertiesRef.
GeluciresGelucires are polyethylene glycol glycerides made up of mono-, di-, and triglycerides, as well as mono- and diesters of polyethylene glycol. [50]
TranscutolProtic solvent, faint odor, colorless limpid liquid, hygroscopic having viscosity 4.1 mPa.s at 25 °C; have exceptional solubilizing capacity due to an alcohol and ether function; Used for skin penetration enhancement [51,52]
PhosphatidylcholineCholine is head-group of phosphatidylcholine and is attached to glycerol of fatty acids via ester-bound to backbone [53]
Poly-lactic acid-co-glycolic acid It is made up of glycolic acid (hydroxy acetic acid) and lactic acid (α-hydroxy propanoic acid) and due to its biocompatible and biodegradable nature, this polymer has been extensively utilized in drug delivery system with superiour loading efficiency [54]
Cholesterol 27-carbon molecule which has amphiphilic nature and contains hydroxyl group linked with phospholipids by hydrogen bonds with the help of flexible carbohydrate linked with bulky steroid ring. During preparation of liposomes, cholesterol avoids aggregation of liposomes and enhances physical stability of membrane of liposomal vesicles and have tendency to generate stable vesicles with high drug loading capacity. [55,56]
Chitosan It is produced via N-deacetylation of chitin. It is biocompatible, and biodegradable polymer of natural origin, therefore considered as harmless substance for use as carrier in production of drug delivery system with high drug loading and entrapment efficiency of several drug molecules [57]
Egg lecithinProvides better stabilizing and encapsulation efficiency; better drug loading [58]
Table 5. Recent studies in emulsion drug delivery system for psoriasis treatment.
Table 5. Recent studies in emulsion drug delivery system for psoriasis treatment.
Drug (Delivery System)ExcipientsPreparation TechniqueClinical Significance and OutcomesRef.
Hydrocortisone
(Multiple emulsion)
Glycerol sorbitan fatty acid ester, Liquid paraffinOil-water-oil emulsificationProlonged topical release of hydrocortisone in epidermis and dermis and the absorbed percentage of hydrocortisone was 1.5-fold greater from the simple emulsion compared to multiple emulsion [43]
Cyclosporine (Micro-emulsion) Oleic acid, Tween 80, Water, Vitamin E-TPGS, Transcutol, Propylene glycolEmulsification
Quick skin uptake and superior skin concentrations achieved after 2 h of contact [44]
Salicylic acid (Micro-emulsion) Polyethylene glycol, Tween 20, Isopropyl myristateO/W emulsificationME with 10% SA does not show any change in storage stability after 6 months except decrease in viscosity after 1 month [45]
8-Methoxsalen (Micro-emulsion)Octanediol, Isopropyl myristate, Tween 80, Span 80, WaterO/W emulsificationThe skin accumulation of 8-Methoxsalen was enhanced 1.5–4.5-fold [46]
Methoxsalen (Micro-emulsion)
Egg phosphatidyl-choline, Chitosan, Ethanol, Acetic acid, Soya oilEmulsification
Methoxsalen loaded chitosan-coated ME show controlled release of drug i.e., 18.75% lesser release than the ME with high drug retention into skin [59]
Methotrexate (Nano- emulsion) Tween 80, Water, Chaulmoogra oilSelf-emulsificationShowed negligible skin irritation and increased penetration into the skin [60]
Clobitasol Propionate &
Calcipotriol (Nano- emulsion)
Cremophor RH 40, Capmul MCM C8 EP, Labrafil® M 1944 CS, WaterOil in water emulsificationThe
nanoemulsion globules of size <100 nm also contributes to improved skin penetration, permeation and retention of drug in deep skin layers
[61]
Table 6. Recent studies in vesicular drug delivery system for psoriasis treatment.
Table 6. Recent studies in vesicular drug delivery system for psoriasis treatment.
Drug (Delivery System)ExcipientsPreparation TechniqueClinical Significance and OutcomesRef.
Tacrolimus and
Curcumin (Liposphere)
Egg lecithin, Tricaprin
tween 80, cremophor RH40, Isopropyl alcohol
Single emulsion solvent evaporationExhibited slow release of drugs and improvement in phenotypic/histopathological features of psoriatic skin [71]
Cyclosporine (Cationic liposome) N-(1-(2,3-dioleoyloxy) propyl)- cholesterol, chloroform, ethanolEthanol injection, thin film hydration, reverse phase evaporation1.67 times rise in the levels of IL-17 on application of IMQ as compared to normal group and showed shear thinning behavior and highly effective and facilitate in psoriasis treatment [72]
Methotrexate (Ethosome)Soya phosphatidyl-choline, chloroform, methanol, hydro-ethanolic solutionMechanical dispersion
Cast film
Provided improved transdermal flux and reduced lag time of 0.9 h across human cadaver skin [73]
Dithranol (Liposome, Niosome)Phosphatidyl choline, cholesterol, span 60, chloroformThin-film hydrationDrug leakage study carried out at 4–8, 25 ± 2 and 37 °C for a period of two months and results revealed that leakage increased at a higher temperature and enhanced permeation with vesicles as signified through flux of dithranol [74]
Tacrolimus (Transferosome)Lipoid E80, Tween 80, Span 80, Dehydrated alcoholThin film hydrationIn vitro drug release was higher in TFs-gel after 24 h in comparision to commercial ointment and from TFs-gel cumulative drug release after 12 h in vitro was 37.6%. and efficient skin target for topical delivery of tacrolimus [75]
Betamethasone dipropionate (Transferosome)Soya phosphatidyl-choline, Sodium deoxycholate, Tween 80, chloroformFilm hydration techniqueThe vesiclecs have 90.19% EE, and have great stability at 25 °C and 4 °C for 6 months and showed significant clinical improvement along with a considerable boost in safety and tolerability [76]
Table 7. Recent studies in particulate drug delivery systems for psoriasis treatment.
Table 7. Recent studies in particulate drug delivery systems for psoriasis treatment.
Drug (Delivery System)ExcipientsPreparation TechniqueClinical Significance and OutcomesRef.
Methotrexate (SLN)Cetyl palmitate, Polysorbate 80Ultra-sonicationIn-vitro results showed a sustained release for 8 h and enhanced skin deposition for effectual treatment of psoriasis [87]
Fluocinolone acetonide (SLN)Compritol 888 ATO, Soya lecithin, Poloxamer 188Modified emulsification ultrasonicationStability results show that SLNs were stable at 4 °C for 3 months and is a promising modality for psoriasis treatment [88]
Diflucortolone valerate (SLN)Geleol, Precirol ATO5, Tristearin, Compritol 888ATO, Poloxamer 407High shear homogenizationTO produce SLNs semisolid preparation 10–20% w/w solid lipid is enough and lipid based surfactants incorporation increased entrapment efficiency and enhanced drug’s solubility [89]
Cyclosporine and calcipotriol (SLN and NLC)Compritol 888 ATO, Precirol, Behenic acid, Gellucire 44/14, Span 20, Cremophor RH-40, Tween 80Hot melt homogenization Deeper and confined drug penetration in epidermal layers for superior psoriatic management [90]
Fluocinolone acetonide (NLC)Compritol, Miglyol 812, Polysorbate 80High speed homogenizationStability findings (3 months) revealed 1.77% and 5.66% percent alteration in EE and particle size and respectively and is regarded higher potential system for psoriasis treatment [91]
Methotrexate (NLC)Witepsol, oleic acid, polysorbate 60, polysorbate 80High-shear homogenization Provided higher drug fluxes of 0.88 μg/cm 2 /h in comparison to free drug (0.59 μg/cm 2 /h) flux [92]
Dithranol (Dendrimer)Poly (amido) amine, ethyl cellulose, carbopol 934, Polyvinyl alcoholQuasi-emulsion solvent diffusionThe results revealed that EE of preparation was in between 71.33% to 49.21%, particle size 28 ± 1.12 mm to 130 ± 1.01 mm and %age yield 66.28% and the formulation produced prolonged efficacy without causing skin toxicities [93]
Dithranol (Dendrimer)EthylenediamineDivergent methodPrimary irritation index of DIT–PPI was revealed to be 1 that means DIT-PPU causes less irritation and have high drug penetration in controlled manner [94]
Dexa-methasone (Nano-crystal) Polyvinyl alcohol, Sodium lauryl sulphateWet bead millingSuperior drug penetration and distribution within skin with reduced dose [95]
Clobetasol propionate (Polymeric microsphere)Poly (d,l-lactide co-glycolide), polyvinyl alcoholSolvent evaporationF8-coded preparation fabricated using PLGA 50:50 at 1:5 drug/polymer ratio and homogenised for 1 min at rpm 8000 was considered as the best preparation and having superior drug efficacy in topical applications [96]
Apremilast (Polymeric nano-particle)Poly (d,l-lactide co-glycolide), Polyvinyl alcoholSingle emulsion solvent evaporation2.25 folds increment in bio-availability of F3 nanoparticles than normal APM suspension and enhancement in half-life and mean residence time leads to long-term retention of nano-particles to provide once-daily regimen [97]
Methotrexate (Gold nano-particle)3-mercapto-1-propansulfonate, Diethylaminoethanethiol hydrochloride, tetrachloroauric (III) acid, sodium borohydrideBioconjugation and functionalizationInduced a diminution of keratinocytes hyper-proliferation, epidermal thickness as well as inflammatory infiltrate in imiquimod-induced psoriasis like mice model [98]
Mometasone furoate (SLN) Glycerol mono-stearate, Tefose-63, Tween-80 Solvent injection method2.67 times more skin deposition as compared to marketed cream and 20 times more in comparsion to plain drug loaded gel and is promising for topical delivery of corticosteroid [99]
Acitretin (NLC) Precirol ATO5, oleic acid, tween 80, tetrahydrofuran Solvent diffusion methodIncrease in Acitretin deposition was found in cadaver skin from ActNLC gel (81.38 ± 1.23%) than to Act plain gel (47.28 ± 1.02%) and enhancement in therapeutic effect for psoriasis and decrease in local side effects [100]
Table 8. Recent works in herbal colloidal drug delivery systems for psoriasis management.
Table 8. Recent works in herbal colloidal drug delivery systems for psoriasis management.
Herbal Constituent (Delivery System)ExcipientsPreparation TechniqueClinical Significance and OutcomesRef.
Celastrol isolated from Tripterygium regelii (Niosome)Cholesterol, carbopol 934, span 20, span 60Thin film hydrationThe developed nanoparticle has particle size of 147 nm and yield of up to 90% and increased water solubility and permeation of celastrol into skin which enhanced its anti-psoriasis activity in mice [123]
Mangiferin isolated from leaves/bark of Mangifera indica (Nano-emulsion)Lipoid® S75, hylouronic acid, Polysorbate 80Ultra-sonicationNanoemulsions having mangiferin significantly reduce oedema ∼20-fold higher than empty nanoemulsions and reduce leucocyte infiltration and showed an anti-inflammatory activity [124]
Acitretin and aloe-emodin Aloe-emodin isolated from plant of genus Aloe (Polymeric nanoparticle)ChitinCentrifugationRevealed greater skin permeation and drug retention in deeper layer of skin with and improve compatibility [125]
Tea tree oil isolated from leaves of Melaleuca alternifolia (Micro-emulsion) Tween 80 EmulsificationShowed superior drug solubilization and bioavailability for topical applications of anti-psoriatic active moieties and bio-actives [126]
Curcumin (Nano-hydrogel)Curcumin, choline-calix[4]arene amphiphileSupra-molecular nano-hydrogelExhibited no significant toxicity and showed effective anti-psoriatic activity in an IMQ-induced psoriasis mouse via decreased pro-inflammation [127]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Singh, S.; Sharma, N.; Behl, T.; Sarkar, B.C.; Saha, H.R.; Garg, K.; Singh, S.K.; Arora, S.; Amran, M.S.; Abdellatif, A.A.H.; et al. Promising Strategies of Colloidal Drug Delivery-Based Approaches in Psoriasis Management. Pharmaceutics 2021, 13, 1978. https://doi.org/10.3390/pharmaceutics13111978

AMA Style

Singh S, Sharma N, Behl T, Sarkar BC, Saha HR, Garg K, Singh SK, Arora S, Amran MS, Abdellatif AAH, et al. Promising Strategies of Colloidal Drug Delivery-Based Approaches in Psoriasis Management. Pharmaceutics. 2021; 13(11):1978. https://doi.org/10.3390/pharmaceutics13111978

Chicago/Turabian Style

Singh, Sukhbir, Neelam Sharma, Tapan Behl, Bidhan Chandra Sarkar, Hasi Rani Saha, Kanika Garg, Supriya Kamari Singh, Sandeep Arora, Md. Shah Amran, Ahmed A. H. Abdellatif, and et al. 2021. "Promising Strategies of Colloidal Drug Delivery-Based Approaches in Psoriasis Management" Pharmaceutics 13, no. 11: 1978. https://doi.org/10.3390/pharmaceutics13111978

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop