Next Article in Journal
Gasdermin D Is a Novel Prognostic Biomarker and Relates to TMZ Response in Glioblastoma
Next Article in Special Issue
Role and Merits of Green Based Nanocarriers in Cancer Treatment
Previous Article in Journal
Experimental Validation of the MRcollar: An MR Compatible Applicator for Deep Heating in the Head and Neck Region
Previous Article in Special Issue
A Systematic Review of Biosynthesized Metallic Nanoparticles as a Promising Anti-Cancer-Strategy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lipid Nanocarriers for Hyperproliferative Skin Diseases

by
Eliana B. Souto
1,*,
Ana L. R. de Souza
2,
Fernanda K. dos Santos
2,
Elena Sanchez-Lopez
3,4,
Amanda Cano
3,4,
Aleksandra Zielińska
5,
Rafał Staszewski
6,
Jacek Karczewski
7,8,*,
Maria P. D. Gremião
2 and
Marlus Chorilli
2
1
CEB—Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
2
Faculty of Pharmaceutical Sciences, São Paulo State University (UNESP), Campus of Araraquara, Rodovia Araraquara Jaú, Araraquara 14800-903, SP, Brazil
3
Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08007 Barcelona, Spain
4
Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08007 Barcelona, Spain
5
Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland
6
Department of Hypertension Angiology and Internal Medicine, Poznań University of Medical Sciences, 61-701 Poznań, Poland
7
Department of Environmental Medicine, Poznań University of Medical Sciences, 61-701 Poznań, Poland
8
Department of Gastroenterology, Dietetics and Internal Diseases, H. Swiecicki University Hospital, Poznań University of Medical Sciences, 60-355 Poznań, Poland
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(22), 5619; https://doi.org/10.3390/cancers13225619
Submission received: 6 October 2021 / Revised: 5 November 2021 / Accepted: 5 November 2021 / Published: 10 November 2021

Abstract

:

Simple Summary

Different drugs, including antiproliferative and corticosteroids in general, are recommended for the treatment of hyperproliferative skin diseases (HSD). The effectiveness of many of these drugs is limited due to their low solubility in water and low penetration in the skin. The loading of these drugs in lipid nanocarriers, such as lipid nanoparticles and liposomes, has been considered as a successful solution to improve the drug bioavailability through the skin, to control their release kinetics and thus reduce the risk of potential side effects. In this work, we discuss the use of lipid nanocarriers loading drugs against HSD.

Abstract

Hyperproliferative skin diseases (HSD) are a group of diseases that include cancers, pre-cancerous lesions and diseases of unknown etiology that present different skin manifestations in terms of the degree and distribution of the injuries. Anti-proliferative agents used to treat these diseases are so diverse, including 5-aminolevulinic acid, 5-fluorouracil, imiquimod, methotrexate, paclitaxel, podophyllotoxin, realgar, and corticosteroids in general. These drugs usually have low aqueous solubility, which consequently decreases skin permeation. Thus, their incorporation in lipid nanocarriers has been proposed with the main objective to increase the effectiveness of topical treatment and reduce side effects. This manuscript aims to describe the advantages of using lipid nanoparticles and liposomes that can be used to load diversity of chemically different drugs for the treatment of HSD.

1. Introduction

Hyperproliferative skin diseases (HSD) do not include only the various types of cancers, as previously thought. They also relate to precancerous lesions and diseases of unknown etiology that can widely vary in the degree and distribution of the skin injuries, such as psoriasis, which can be caused by inflammatory responses [1,2]. Among the most commonly used drugs in the treatment of HSD, anti-proliferative agents, such as 5-aminolevulinic acid, 5-fluorouracil, paclitaxel, podophyllotoxin, methotrexate, imiquimod, and corticosteroids stand out [3].
The main limitations of these drugs are related to their physicochemical properties that hinder permeation through the skin [4,5,6]. The use of lipid nanocarriers (e.g., solid lipid nanoparticles (SLN), nanostructured lipid carriers (NLC), and liposomes), has been described as a successful approach to formulate these drugs (Figure 1) for the treatment of HSD to increase the efficacy of the topical treatment, in addition to reducing side effects [5,7].
The use of lipid carriers for the incorporation of drugs is not a recent approach. In 1965, Gregoriadis proposed for the first time the use of phospholipids in form of liposomes for drug delivery [8]. Amphiphilic molecules, as phospholipids, have a polar region (ionic) and an apolar region [9,10]. When present at a concentration higher than their Critical Micellar Concentration (CMC) and excess of water, can form different types of supramolecular aggregates, such as micelles, monolayers, multilayers and liposomes [9,11].
Liposomes are made up of one or more spherically arranged bilayers, separated by aqueous phases, encompassing an internal aqueous compartment [11,12]. These vesicles are organized in the presence of water, and, in part, the bilayer orientation is determined by the nature of the polar groups and carbon chains. Various amphiphilic compounds, containing two carbon chains, including the natural phospholipids and synthetic compounds [9,10], can be used as structural elements of liposomes while the production method can be fine-tuned to control the size and morphology of the vesicles [11].
Liposomes can be classified into three main groups, namely, multilamellar liposomes (MLL), small unilamellar liposomes (SUL), and large unilamellar liposomes (LUL) [12]. The MLL is formed by several concentric bilayers, interspersed with aqueous compartments. Their diameter is variable according to the number of lamellae, ranging approx. from 400 to 3500 nm. SUL, with a diameter <50 nm, consists of only a bilayer and a small watery compartment. LUL are also made up of only one bilayer, but with a large aqueous cavity. Their diameter ranges approx. from 200 to 1000 nm. Liposomes vary in size and homogeneity according to the production method and can be prepared by simply dispersing a film of amphiphilic molecules with mechanical agitation, sonication, reverse phase evaporation, extrusion, among others [13].
Most phospholipids do not form SUL spontaneously, requiring the supply of energy, for example, sonication [13]. During sonication, membrane fragments are formed, in which a hydrophobic part is exposed to the polar aqueous medium. When there is a transfer from an apolar medium to an aqueous medium, entropy is unfavorable. To overcome this, hydrophobic fragments join together. The unfavorable entropy of the interaction of the hydrophobic part of the fragments is equivalent to the unfavorable energy of the packaging. As a result, liposomes have a small radius of curvature surface. When energy is balanced, smaller liposomes are formed [14].
Liposomes are used as carriers for different molecules, offering a therapeutic improvement to loaded drugs, due to the high degree of biocompatibility and biphasic character [15]. The versatility of liposomes makes them suitable carriers for highly hydrophilic, highly lipophilic, and amphiphilic molecules.
The interaction of lipid nanocarriers with the skin is mainly governed by the nanocarriers’ structure which is defined by their lipid and surfactant composition. Potential alterations in the lipid domains of the stratum corneum were described for liposomes [16], whereas lipid nanocarriers with a solid matrix (e.g., solid lipid nanoparticles, nanostructured lipid carriers) are described to adhere onto the skin surface creating a lipid film that promotes drug permeation by increasing skin hydration [17]. The degree of drug permeation on the skin is, however, a matter of concern, whether a superficial (topical) or a deeper effect is expected without reaching systemic circulation (without transdermal delivery). The size and lipid composition are instrumental to control the extension of drug permeation in the skin. The presence of lipids able to disrupt the epidermis will promote a greater drug permeation [18].
Highly hydrophilic drugs or active pharmaceutical ingredients (APIs) are encapsulated in liposomes dissolved in the inner aqueous core. Lipophilic APIs will be encapsulated within the phospholipid bilayer, whereas amphiphilic APIs will be placed in between the aqueous core and lipid bilayers governed by their partition coefficient [15].
SLNs are considered promising carriers to modify the release profile of loaded APIs for the topical administration of drugs [19,20,21,22,23,24,25]. SLNs have a matrix composed of lipids that are solid at room and body temperature [17,26,27], being successfully used to improve the bioavailability of APIs by different administration routes, such as dermal, ocular, pulmonary, topical, among others [19,28,29]. SLNs have, however, some characteristics that limit their use, such as the low loading capacity due to the risk of expulsion of APIs from the matrix attributed to polymorphic changes of lipids during shelf-life, and also the high amount of water in the dispersions, which can vary from 70% to 90% [28,29,30].
To overcome the limitations of SLN, NLC has been developed [31,32]. The NLC matrix is composed of a mixture of a solid lipid and a liquid lipid that also melts above room and body temperatures thereby allowing to modify the release profile of loaded drugs [7,26,30,32]. The risk of polymorphic changes in the NLC matrix is reduced by the contribution of the liquid lipid, promoting a higher loading capacity when compared to SLN. Table 1 summarizes the main lipid nanocarriers that can be used in the treatment of HSD. Over the last few years, many studies have been published focusing on the topical application of lipid nanocarriers, highlighting their ability to load chemically different drugs to improve the bioavailability through the skin, by delivering them via controlled release kinetics, and showing no or low risk of systemic side effects or cytotoxicity [33,34]. Besides, lipid nanocarriers applied topically improve skin hydration attributed to their occlusive effects. Moisturized skin may also contribute to improved skin permeability of loaded drugs [35,36]. In this work, we discuss the background of HSD, such as basal cell and squamous cell carcinomas, skin melanoma and psoriasis, and how lipid nanocarriers can be an alternative over conventional treatments to improve drug bioavailability and reduce side effects. The selection of the lipid nanocarrier used in HSD is governed by the type of drug to be loaded (lipophilic versus hydrophilic), as these carriers are produced from biocompatible, biodegradable and biotolerable lipids that do not pose toxicological problems [33,34]. This aspect is instrumental for particles not only used in systemic treatments but also in topical treatments. While the purpose of using lipid nanocarriers to treat HSD is to increase the drug permeation/penetration into deeper skin layers, which is facilitated in diseased skin with compromised stratum corneum, for other topical applications (e.g., cosmetics) the drug has to remain onto healthy skin with minimal risk of permeating the skin.

2. Hyperproliferative Skin Diseases

2.1. Skin Cancer

Skin cancer is the general term used to refer malignant neoplasms which themselves refer to skin cells that have undergone a transformation and have multiplied in a disordered and abnormal way giving rise to a new tissue (neoplasia). Carcinoma refers to a cancer that begins in the epithelial tissue and can spread to other parts of the body. In the skin, one can describe the basal cell carcinoma, which is a slow-growing cancer that rarely spreads to other parts of the body. Squamous cell carcinoma also rarely spreads, but it occurs more frequently than basal cell carcinoma. These two types of carcinomas are sometimes called non-melanoma skin cancers. Another type of cancer that occurs on the skin is melanoma, which starts in the melanocytes, and all types of melanomas have the potential to cause metastasis if not previously treated [37].
The classification of a tumor as benign versus malignant is based on its capacity to originate metastases. Benign carcinoma is usually isolated, presenting as stable nodules, whereas the malignant lesion tends to invade other tissues, it grows rapidly and causes lesions with metastases. With the aging of the population, there is a simultaneous increase in actinic keratosis (pre-cancer lesions) and non-melanoma skin cancer, with solar radiation as the main cause. The cumulative effect of the sun can damage skin cells and lead to the development of skin cancer [37]. The oncogenic effects of solar radiation result from the action of electromagnetic waves on deoxyribonucleic acid (DNA). The formation of pyrimidine dimers in DNA by ultraviolet (UV) light, if not repaired, promotes errors in transcription that lead to the appearance of cancer.
UV radiation is primarily responsible for the development of skin cancer. Every time the skin is exposed to this radiation, either by direct sun exposure or through the use of artificial tanning techniques, lesions occur in the deeper layers of the skin that are not repaired. These lesions accumulate and, with systematic exposure to UV rays, they can end up producing malignant mutations, especially in people with fair skin. The keratinocytes of these patients are damaged by UV light, but the cells repair a large part of the DNA damage and the carcinoma only develops later. Thus, preventive measures of this disease from an early age, through the constant and rigorous use of photoprotection, is instrumental to reduce the risk of skin cancer [38].
In black people, much of this radiation is absorbed by the pigment melanin, not reaching the keratinocyte target; therefore, black skin does not normally develop basal cell and squamous cell carcinomas.

2.1.1. Basal Cell Carcinoma

Basal cell carcinoma (basalioma or basal cell epithelioma) is the most common skin cancer, representing about 70% of all types. It originates in the cells of the basal layer of the epidermis and has a direct relationship with skin exposure to solar radiation, mainly to UVB radiation.
Basal cell carcinomas generally occur in individuals aged from 17 to 35 years [39]. They affect individuals with light skin, hair, and light eyes (Caucasians), mainly rural workers, and redheads and individuals with “freckles”. It usually appears after the 4th decade of life and is preferentially located in the areas of the body most exposed to the sun, such as face, neck, and back. Therefore, sun protection is the best way to prevent its appearance [38]. This tumor appears initially as reddish lesions on the skin that peel off and are slightly hardened. Its growth is very slow and gradual, not promoting metastases; however, they can turn into wounds that do not heal.
The vast majority of injuries appear on the face and in individuals over 40 years old. Its most typical presentation begins as a small, consistent lesion, pink or translucent in color and “pearly”, smooth and shiny, with thin blood vessels on the surface and grows progressively and slowly, giving rise to a nodule with subsequent central ulceration covered with a crust, which, when removed, leads to bleeding.
The treatment of basal cell carcinoma in the initial phase usually results in a 95% success rate of cure, being done through surgery (excision) or laser removal (cauterization). If it progresses without treatment, it can become very aggressive, invading the surrounding tissues and causing major defects and mutilations, especially in certain anatomical areas (nose, pinna, eyelids, etc.).

2.1.2. Squamous Cell Carcinoma

Squamous cell carcinoma (squamous, squamous, or planocellular epithelioma) is a malignant tumor of the skin, characterized by variation in size and shape, hyper-colored and/or giant nuclei, loss of the nucleus-cytoplasm and nucleus-nucleolus relationship, frequently presenting mitoses. In the epithelium, the arrangement of cells is irregular and successive layers are not formed from the basal layer to the corneal layer; cornification is disordered and appears abruptly, off the surface and without normal progressive transformation.
Squamous cell carcinoma results from the atypical proliferation of spinous cells, of an invasive character, which can produce metastases. In addition, it usually appears after the age of 50. The characteristic lesions are nodules or plaques, reddish and thick, which bleed at the slightest trauma. The evolution of this carcinoma is fast, characterized by an increase in the size of these nodules, which may become verrucous or vegetating. Occasionally, they may develop lesions with high edges and a crusted surface on a high granular base.
This type of cancer can appear in areas of healthy skin or previously compromised by some other process, such as scars from old burns, chronic wounds, or injuries due to the skin exposure to solar radiation, such as actinic keratoses, which are caused by excessive accumulation of keratin in the epidermis [39]. Squamous cell carcinoma develops faster than basal cell carcinoma, reaching the skin and mucous membranes (oral mucosa and external genitalia) and can metastasize to other organs if not treated early.
The etiology of squamous cell carcinoma is multifactorial including, in addition to intense sun exposure, the use of immunodepressants (drugs used to prevent organ rejection after transplantation), exposure to industrial carcinogens (tar and oils), tobacco, among others [38]. The treatment of this carcinoma is usually surgical, through the total removal of the lesion (with control of the margins) and it must be carried out as early as possible to avoid the occurrence of metastases. Radiotherapy is also effective, especially when the location is in the lip.

2.1.3. Melanoma

Melanoma consists of atypical melanocytes of the basal layer of the skin, with an invasive character that may result in metastasis to other regions of the body [40]. In addition, melanoma is most often located on the back, upper and lower limbs. It can appear spontaneously or can originate from a pre-existing signal [41].
Clinically, malignant skin melanoma is often asymptomatic, but the itching and bleeding wounds may represent the first manifestations of melanoma. Therefore, the most important clinical sign of a possible malignant degeneration is the color change in a pigmented lesion.
Many APIs have been proposed for the treatment of melanoma, but only a few show activity in more than 10% of patients [42]. The use of cytotoxic APIs remains the main form of chemotherapy for cancer. Cytotoxic APIs belong to a diverse class of compounds effective mainly because they are toxic to cells that are growing and dividing rapidly [43]. Figure 2 shows a schematic representation of superficial spreading melanoma and possible pathways of skin penetration of nanoparticles.

2.1.4. Applications of Lipid Nanocarriers in Skin Cancer

Lipid nanocarriers have been proposed either for the protection of the skin from sun exposure (due to their inherent UV scattering effect) or for the delivery of chemotherapeutic drugs by topical administration [43,44].
5-fluorouracil (5-FLU), synthesized by Heidelberg et al. in 1957, is one of the oldest and most used chemotherapeutic drugs in the treatment of cancer [45]. It is an anti-metabolite of low molecular weight and easily soluble in water [43]. The effectiveness of 5-FLU was firstly described for the treatment of cancer whose lesions improved after systemic treatment with these APIs. Similar results were observed with its topical application in the treatment of actinic keratosis. Studies have shown that treatment with 5% 5-FLU improved lesions by 70% to 75% [45,46,47,48].
The photodynamic therapy (PDT) for the targeting of photosensitive drugs, such as 5-aminolevulinic acid (5-ALA), which is used to induce the production of protoporphyrin IX in cells [5,49,50], has also been exploited for the treatment of skin cancer. PDT is based on the use of a photochemical reaction between a light source of a specific wavelength and a photosensitizer in the presence of oxygen [51]. It is effective, safe, and has a long-term effect; however, because it has a hydrophilic precursor, it has a limited permeation through the skin.
PDT has been proposed in combination with lipid nanocarriers to overcome this limitation. Fang et al. described the degree of cutaneous penetration of 5-ALA in liposomes [52]. The authors observed that when encapsulated in liposomes, 5-ALA increased the production of protoporphyrin in the skin. Fadeel et al. proposed the use of polyethylene glycol-grafted (PEGylated) lipid nanocarriers loaded with curcumin to target skin cancer by photodynamic therapy [51]. Curcumin is an aromatic phytoextract obtained from the turmeric rhizome of Curcuma longa, with known anti-oxidant, anti-inflammatory and anti-cancer properties [53]. The poorly water-solubility of curcumin, however, limits its bioavailability in vivo. The loading of curcumin into lipid nanocarriers has been shown to improve the therapeutic effect of this polyphenol [54,55]. The human skin cancer cell line A431 was used to test the dark and photo-cytotoxicity of the curcumin-loaded PEGylated lipid nanocarriers, showing enhanced viability when cells were treated with the loaded curcumin when compared with the drug suspension [51]. By confocal laser microscopy, the in vivo studies in mice confirmed the increased permeation of curcumin, while histopathological analysis confirmed the presence of the drug in deeper layers of the skin after irradiation [51].
Palliyage et al. described the development of negatively charged solid lipid nanoparticles (SLN) for the dual loading of curcumin and resveratrol for the inhibition of proliferation of highly aggressive melanoma cells with high success [56]. Analysis by electrical cell-substrate impedance sensing suggested the potential blocking effect on cell migration of B16F10 melanoma cells by the dual therapy. A synergistic effect in inhibiting SK-MEL-28 melanoma cell proliferation was also described.
Temoporfin (mTHPC) is a potent second-generation synthetic photosensitizer with an effect on the photodynamic therapy of primary non-melanomatous tumors of the skin, head, and neck after intravenous administration topical formulations of mTHPC on the market is restricted [57]. Therefore, topical formulations containing mTHPC can be interesting in photodynamic therapy for both non-malignant diseases (psoriasis) or malignant skin diseases (basal cell carcinoma). However, mTHPC is a highly lipophilic drug with a molecular weight of 680 Da, which makes its skin permeation very low.
Dragicevic-Curic et al. described that deposition in the stratum corneum and accumulation in the deeper layers of the skin of mTHPC were greater for liposomes containing positively charged lipids. mTHPC was released into the stratum corneum and reached deeper skin layers in enough amount for topical PDT by administration of neutral or surface-loaded liposomes.

2.2. Psoriasis

Psoriasis is a chronic inflammatory disease of the skin and joints, autoimmune, with great polymorphism of clinical expression [58]. This disease is one of the most frequently described dermatoses in clinical practice, affecting men and women in the same way, characterized by cell renewal seven times faster than for normal skin, which is 28 days [7,59]. This disease can manifest itself in several different ways and presents itself in the form of minimal lesions to a more severe form, called erythrodermic psoriasis, in which the skin of the entire body may be compromised. However, the most frequent form of presentation is plaque psoriasis, characterized by the appearance of well-defined erythematous squamous skin lesions with chronic evolution. These scales are usually whitish and are most often located on the elbows, knees, scalp, and torso. Psoriasis lesions usually have no symptoms, but there may be mild itching at the site. When plaques regress, they usually leave a lighter area of skin in the affected area [60]. A metadata analysis published in 2019 reported the association between psoriasis and the risk of developing cancer [61].
Less common presentations are nail psoriasis, with lesions only on the nails, pustular psoriasis, with the formation of pustules mainly on the palms and soles of the feet, and psoriatic arthritis, which is characterized by joint inflammation that can even cause total destruction of the joint, usually affecting the fingers. The degrees of severity of psoriasis are divided into mild, moderate, and severe, one of the most serious being called inverse psoriasis, where the lesions are flat, inflamed, reaching large areas [62].
For some time, the question remained about the etiology of this pathology, whether it was just dermatological or immunological [63,64,65]. In this way, two possible theories have been proposed. The first assumes an unknown primary defect in keratinocytes that would produce cytokines if activated directly by physical or chemical trauma. These substances would activate T cells to release cytokines, with the consequent proliferation of lymphocytes and keratinocytes in the epidermis. The second theory deals with a mechanism secondary to the persistent activation of T lymphocytes by specific exogenous or endogenous factors (antigens, superantigens, or autoimmunity), leading to the proliferation and abnormal differentiation of keratinocytes [7,63].
The therapeutic strategy to be used in the treatment of psoriasis varies according to the conditions of each patient. The aim of the treatment is to decrease epidermal proliferation and the underlying inflammation, as there is no cure for this disease. Systemic treatment consists of using APIs such as cyclosporine, acitretin, and methotrexate, which can usually lead to hepatotoxicity and myelotoxicity. For topical therapy, the available treatment consists of the use of corticosteroids, calcipotriene, phototherapy with ultraviolet B radiation (UVB), among others [66,67].
The administration of drugs directly on the skin lesion makes it possible to minimize side effects on other organs and in the uninjured skin. In mild forms of psoriasis, topical therapy is usually enough to control the lesions. In moderate to severe forms, the association of local treatment with phototherapy and/or systemic therapy provides more comfort to the patient and accelerates skin recovery [7].
It is observed, therefore, that the APIs used in the treatment have serious side effects and research on new APIs for this purpose is limited. It is known that introducing a new active ingredient in the market, in addition to taking several years of research, involves very high costs. The alternative used to circumvent these high costs is to resort to the strategy of developing delivery systems for these active ingredients, which has allowed for an increase in their efficiency, the re-introduction of other active ingredients previously discarded for their undesirable properties, and the improvement of new substances before they are actually launched on the market or used in therapy [68].
Thus, in recent years, new drug delivery systems have been developed, such as lipid nanocarriers, with the objective not only of increasing the selectivity and effectiveness of active ingredients, but also allowing the reduction of its total necessary dose, minimizing toxic side effects, and allowing control of drug release [69,70].

Lipid Nanocarriers Applied in the Treatment of Psoriasis

Trotta et al. evaluated the permeation of methotrexate administered in phosphatidylcholine (PC) liposomes, hydrogenated phosphatidylcholine (HPC) and in deformable liposomes—PC or HPC liposomes containing a compound with emulsifying properties, dipotassium glycyrrhizinate—using dissected ears and hydrated pigs, maintaining the transepidermal hydration gradient [71]. The authors observed that the amount of methotrexate that permeated pig skin after a 24-h period was similar for the drug in aqueous solution or encapsulated in PC or HPC liposomes. However, when methotrexate was delivered in deformable liposomes, there was an increase in the amount of permeated drug. Drug penetration through the stratum corneum was facilitated when using deformable liposomes under the influence of transcutaneous hydration. The higher methotrexate permeation was attributed to the fusion of the deformable liposomes with the skin, facilitated by the increase in the fluidity of the phospholipid bilayers containing dipotassium glycyrrhizinate.
Another class of drugs used in the treatment of psoriasis is glucocorticoids [72]. Glucocorticoid receptors are located on viable keratinocytes and fibroblasts of the epidermis and dermis [73,74]. However, to reach these cells, drugs have to permeate the stratum corneum. The anti-proliferative and anthropogenic effects of steroids are due to the inhibition of IL-1α in fibroblasts; the anti-inflammatory effect results from the cytokine’s inhibition in keratinocytes [6,7,72].
Sivaramakrishnan et al. produced SLN by high-pressure homogenization, consisting of glyceryl behenate and poloxamer 188 (SLN A) or glyceryl and polysorbate 80 palmitoestearate (SLN B), for incorporation of 0.1% betamethasone valerate (BMV) [75]. The in vivo skin penetration studies of the different formulations (SLN A, SLN B, conventional cream containing 0.1% BMV, and nanoemulsion containing medium-chain triglyceride and poloxamer 188) were performed on the abdominal or breast skin of healthy humans. There was a fourfold increase in the penetration of BMV incorporated in SLN A compared to the control (cream with 0.1% BMV), which was not observed for BMV incorporated in SLN B and in nanoemulsions.
Zhang and Smith (2011) developed SLN containing BMV by the solvent injection method using monostearate or beeswax and lecithin as a surfactant as an oil phase [76]. In vitro skin permeation studies were carried out on Franz cells for the different formulations (lotions, suspensions, mono-stearic SLN, and beeswax SLN), in addition to assessing the distribution of corticosteroids and the formation of a reservoir in the skin. Lower BMV permeation flow [J = 0.1547 ± 0.009 µg/(cm2·h)] through the skin and greater APIs reservoir formation were observed for monostearate SLNs than for commercial lotions and APIs suspension, which can be advantageous for minimizing systemic absorption of APIs and, consequently, reducing the side effects that often occur when corticosteroids are absorbed into the bloodstream, in addition to increasing the amount of APIs at the site of action. In addition, the amount of BMV quantified in the epidermis and dermis was the highest when administered in form of monostearate SLN.
Fang et al. loaded 8-methoxypsoralen into Precirol and squalene composed nanostructured lipid carriers and demonstrated that drug entrapment minimized the permeation differentiation between normal and hyperproliferative skin compared to the free drug in nude mice [31].
Triamcinolone is an APIs widely used to relieve the inflammatory symptoms of corticosteroid-responsive dermatitis [77]. Yu and Liao (1996) developed ML and SUL containing triamcinolone and evaluated the effect of liposomal formulations on the skin permeation of these APIs [78]. The results were compared with the commercial triamcinolone ointment. The permeation study was performed on Franz cells using Wistar rat skin. The results obtained showed that the type of liposome (ML or SUL), its charges (positive, negative, or neutral), and the lipid composition influence the amount of APIs that permeates and is retained on the skin. Liposomes facilitated both the retention and cutaneous permeation of APIs. The permeability of triamcinolone was higher for liposomes than for conventional ointment. This in vitro study demonstrated that liposomes could increase the intradermal concentration of poorly water-soluble drugs.
Tretinoin is a metabolite of vitamin A that has also been widely used for the treatment of various inflammatory and proliferative skin diseases (e.g., psoriasis, acne, epidermotropic lymphomas originating from T-cells and epithelial cancer). Tretinoin plays a role in the regulation of epithelial cell growth and differentiation, sebum production and collagen synthesis [79]. However, because of its low water solubility, high instability in contact with air, light and heat, in addition to local skin irritation (risk of erythema, flaking and burning), the topical use of TRE is greatly limited [28].
To overcome these drawbacks, tretinoin has been formulated in delivery systems [80,81,82]. The loading of tretinoin in liposomal formulations reduced the undesirable effects of APIs on the skin’s surface, maximized its accumulation in the skin and prevented fast degradation. In vitro studies have reported an increase in the accumulation of tretinoin in the skin and in vivo studies have shown reduced irritation after liposomal treatment [83].
Acitretin, the second generation of aromatic retinoids, a metabolite of vitamin A, is another drug used in the treatment of psoriasis. This drug has the ability to establish a normal pattern of epithelial cell growth [84,85]. In a clinical study published by Agrawal et al. (2010), acitretin-loaded in NLC significantly reduced erythema compared to the conventional form [84]. The authors also noted a reduction in adverse effects, suggesting that treatment may be indicated primarily for patients who experience adverse reactions to topical psoriasis treatments.
Dithranol is highly effective for the treatment of psoriasis, but it is associated with irritation and necrotizing effects, burning and blemishes both on normal skin and on diseased skin [86]. Another substance reported in the literature is triptolide (diterpenoid triepoxide), a purified compound from traditional Chinese medicine extracted from a vine called Tripterygium wilfondii Hook F. Triptolide is quite efficient when absorbed into the bloodstream for the treatment of inflammatory and autoimmune diseases, and it presents immunosuppressive, antineoplastic and antifertility activities. Because of its low solubility in water and its adverse effects already reported in China, especially at the level of the gastrointestinal tract (e.g., nausea, vomiting, stomach pain, diarrhea, duodenal ulcer and hemorrhages), its clinical use is limited. Thus, the development of new triptolide delivery systems would lead to significant advantages in the clinical use of this drug [87,88].
Mei et al. (2005) developed SLN with triptolide and subsequent incorporation in Carbomer 940 hydrogels [88]. The in vitro skin permeation studies of triptolide from various dispersions of SLN and the hydrogel containing SLN were performed using the abdominal skin of rats with subcutaneous adipose tissue. Hydrogels with smaller particle sizes (tristearin SLN hydrogels) showed a greater amount of accumulated triptolide and prolonged release (during 24 h), in contrast to SLN dispersions, which released a greater amount of drug in the first 8 h. In addition, the percentage of transdermal absorption accumulated in 12 h was 45.3% for the conventional triptolide hydrogel, while for the SLN-triptolide hydrogel it was 73.5%, this percentage being determined by the diffusion of TP a from the solid lipid matrix to the hydrogel and, subsequently, to the skin. The formulation’s acute anti-inflammatory activity was evaluated in vivo in Wistar rats using carrageenan-induced paw edema as a parameter. The results obtained indicated that SLN-triptolide, hydrogels of SLN-triptolide and triptolide alone, showed acute anti-inflammatory activity, and the hydrogel of SLN-triptolide further reduced the paw edema induced by carrageenan when compared with SLN-triptolide and the control group. This anti-inflammatory effect was twice as high as the conventional triptolide hydrogel and especially better than that obtained for the control group, in which sodium diclofenac was used.
Tacrolimus (FK506) is an API with a well-tolerated immunosuppressive effect, extensively studied clinically for the treatment of chronic plaque psoriasis [89]. The most common adverse effects associated with its use include low and variable bioavailability, burning sensation, and itching at the application site, in addition to the potential to increase the risk of skin infections by altering the local immune response [90].
The in vitro permeation studies for tacrolimus were carried out with the use of the abdominal skin of hairless rats in Franz cells. They were performed by comparing the suspension of liposomes and liposomes incorporated in a Carbopol gel with the formulation of tacrolimus in propylene glycol, containing equivalent amounts of drugs [91]. With this study, it was found that there was a significant increase in the permeation of tacrolimus carried in the liposomes compared to the aqueous solution or Carbopol gel. The drugs permeated for 24 h were 65.71% and 61.45% from the liposomal suspension and liposome gel, respectively, while only 49.57% and 35.21% of the APIs permeated in the case of aqueous solution and Carbopol gel, respectively. Higher flow values obtained with the liposome suspension (2.623 ng/cm2/h) and liposome gel (1.740 ng/cm2/h) than those obtained with an aqueous solution (0.932 ng/cm2/h) and Carbopol gel (1237 ng/cm2/h) confirm the effect of APIs encapsulation in liposomes to improve its permeation. The phospholipid-rich domains of the vesicles may have helped to produce the deposition effect of APIs, reflecting a higher amount of drug retention in the skin layers in the case of liposomal formulations.
Photochemotherapy with psoralens is widely used in the treatment of psoriasis. The term PUVA, from the English “Psoralen UVA light therapy”, is used to define a group of therapeutic techniques that use psoralens and UV light. These are photosensitizing compounds derived from tricyclic furocoumarins, that occur naturally in some plants, or are produced by chemical synthesis. These compounds have a strong absorption band in the region of 200–350 nm. The main psoralens used in PUVA are 4,5′, 8-trimethylpsoralen (TMP), 8-methoxypsoralen (8-MOP) and 7-methylpyrido-(3,4-c)-psoralen (7-MOP), in combination with UVA radiation (PUVA therapy), which are used therapeutically in the treatment of proliferative skin disorders such as psoriasis and vitiligo.
Fang et al. developed SLN and NLC containing psoralens and examined the in vitro skin permeation of APIs, using hyperproliferative skin to mimic a clinical situation [52]. The permeation of 8-MOP followed zero-order kinetics for nanoparticulate systems and a biphasic release profile for the aqueous suspension. The flow of drugs through the rats’ skin was higher for NLC. The psoralens permeation increased in the following order: 8-MOP > 7-MOP > TMP.
Regardless of the type of HSD, the selection of the lipid nanocarrier is dependent on the type of drug (lipophilic versus hydrophilic), which will govern the selection of the lipid materials (fatty acids, mono-, di- or triglycerides, phospholipids, etc) in which the drug will show the highest solubility. The higher the solubility in the materials used in the production of the carrier, the higher the loading capacity and encapsulation efficiency. The increased encapsulation parameters will promote the capacity to reduce the dose required for a therapeutic effect and may thus reduce the risk of side effects.
Table 2 summarizes examples of APIs formulated into lipid nanocarriers for the treatment of hyperproliferative diseases.

3. Conclusions

The skin has numerous advantages both for topical and systemic administration of drugs. For the treatment of skin pathologies, such as hyperproliferative skin diseases, nanoparticles play a significant role in retaining the drug at the site of action and by modifying the release profile. The use of lipids resembling those existing in the skin structures in the production of lipid nanocarriers offers the additional advantage of being compatible and of creating a protective lipid film. The increased retention of antiproliferative drugs in the skin without inducing significant and irreversible changes in its barrier function opens the opportunity for developing new, more effective treatments for skin chemotherapy and psoriasis. Studies show that drugs used topically in HSD, when loaded into lipid nanoparticles, have a lower incidence of side effects and, particularly, demonstrate an increase in clinical efficacy. On the other hand, progress in particle engineering, in the design of new nanomaterials for the modified release of drugs, and a better understanding of how these particles interact with the (healthy versus damaged) skin will lead to significant advances in the treatment of hyperproliferative skin diseases.

Author Contributions

Conceptualization, E.B.S., A.L.R.d.S., F.K.d.S., M.P.D.G. and M.C.; writing—original draft preparation, A.L.R.d.S., R.S., J.K., F.K.d.S., A.C., E.S.-L. and A.Z.; writing—review and editing, E.B.S., A.C., E.S.-L., A.Z., M.P.D.G. and M.C.; visualization, A.L.R.d.S.; supervision and funding acquisition, E.B.S., J.K., M.P.D.G. and M.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Portuguese Science and Technology Foundation (FCT/MCT), European Funds (PRODER/COMPETE)—project UIDB/04469/2020 (strategic fund), co-financed by FEDER, under the Partnership Agreement PT2020. The work is also supported by the National Science Centre within the MINIATURA 4 for a single research activity (grant no: 2020/04/X/ST5/00789) and by the START 2021 Program of the Foundation for Polish Science (FNP) granted to Aleksandra Zielińska.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pohla, L.; Ottas, A.; Kaldvee, B.; Abram, K.; Soomets, U.; Zilmer, M.; Reemann, P.; Jaks, V.; Kingo, K. Hyperproliferation is the main driver of metabolomic changes in psoriasis lesional skin. Sci. Rep. 2020, 10, 1–7. [Google Scholar] [CrossRef] [Green Version]
  2. Thatikonda, S.; Pooladanda, V.; Sigalapalli, D.K.; Godugu, C. Piperlongumine regulates epigenetic modulation and alleviates psoriasis-like skin inflammation via inhibition of hyperproliferation and inflammation. Cell Death Dis. 2020, 11, 17–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sahu, S.K.; Raj, R.; Raj, P.M.; Alpana, R. Topical Lipid Based Drug Delivery Systems for Skin Diseases: A Review. Curr. Drug Ther. 2020, 15, 283–298. [Google Scholar] [CrossRef]
  4. Kim, S.C.; Yoon, H.J.; Lee, J.W.; Yu, J.; Park, E.-S.; Chi, S.-C. Investigation of the release behavior of DEHP from infusion sets by paclitaxel-loaded polymeric micelles. Int. J. Pharm. 2005, 293, 303–310. [Google Scholar] [CrossRef] [PubMed]
  5. Prow, T.W.; Grice, J.E.; Lin, L.L.; Faye, R.; Butler, M.; Becker, W.; Wurm, E.M.T.; Yoong, C.; Robertson, T.A.; Soyer, H.P.; et al. Nanoparticles and microparticles for skin drug delivery. Adv. Drug Deliv. Rev. 2011, 63, 470–491. [Google Scholar] [CrossRef]
  6. Zhang, Y.; Li, J.; Lang, M.; Tang, X.; Li, L.; Shen, X. Folate-functionalized nanoparticles for controlled 5-Fluorouracil delivery. J. Colloid Interface Sci. 2011, 354, 202–209. [Google Scholar] [CrossRef]
  7. Kolenyak dos Santos, F.; Helena Oyafuso, M.; Priscila Kiill, C.; Palmira Daflon-Gremiao, M.; Chorilli, M. Nanotechnology-based drug delivery systems for treatment of hyperproliferative skin diseases-a review. Curr. Nanosci. 2013, 9, 159–167. [Google Scholar]
  8. Gregoriadis, G. Engineering liposomes for drug delivery: Progress and problems. Trends Biotechnol. 1995, 13, 527–537. [Google Scholar] [CrossRef]
  9. Fernandes, A.R.; Sanchez-Lopez, E.; Santini, A.; dos Santos, T.; Garcia, M.L.; Silva, A.M.; Souto, E.B. Mono- and Dicationic DABCO/Quinuclidine Composed Nanomaterials for the Loading of Steroidal Drug: 32 Factorial Design and Physicochemical Characterization. Nanomaterials 2021, 11, 2758. [Google Scholar] [CrossRef]
  10. Fernandes, A.R.; dos Santos, T.; Granja, P.L.; Sanchez-Lopez, E.; Santini, A.; Garcia, M.L.; Silva, A.M.; Souto, E.B. DABCO-Customized Nanoemulsions: Characterization, Cell Viability and Genotoxicity in Retinal Pigmented Epithelium and Microglia Cells. Pharmaceutics 2021, 13, 1652. [Google Scholar] [CrossRef]
  11. Teixeira, M.D.C.E.A.; Carbone, C.; Souto, E. Beyond liposomes: Recent advances on lipid based nanostructures for poorly soluble/poorly permeable drug delivery. Prog. Lipid Res. 2017, 68, 1–11. [Google Scholar] [CrossRef] [PubMed]
  12. Souto, E.B.; Macedo, A.S.; Dias-Ferreira, J.; Cano, A.; Zielińska, A.; Matos, C.M. Elastic and Ultradeformable Liposomes for Transdermal Delivery of Active Pharmaceutical Ingredients (APIs). Int. J. Mol. Sci. 2021, 22, 9743. [Google Scholar] [CrossRef] [PubMed]
  13. 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] [PubMed] [Green Version]
  14. Yeagle, P. Modulation of membrane function by cholesterol. Biochimie 1991, 73, 1303–1310. [Google Scholar] [CrossRef]
  15. Gulati, M.; Grover, M.; Singh, S.; Singh, M. Lipophilic drug derivatives in liposomes. Int. J. Pharm. 1998, 165, 129–168. [Google Scholar] [CrossRef]
  16. Kuntsche, J.; Bunjes, H.; Fahr, A.; Pappinen, S.; Rönkkö, S.; Suhonen, M.; Urtti, A. Interaction of lipid nanoparticles with human epidermis and an organotypic cell culture model. Int. J. Pharm. 2008, 354, 180–195. [Google Scholar] [CrossRef]
  17. Souto, E.B.; Baldim, I.; Oliveira, W.P.; Rao, R.; Yadav, N.; Gama, F.M.; Mahant, S. SLN and NLC for topical, dermal, and transdermal drug delivery. Expert Opin. Drug Deliv. 2020, 17, 357–377. [Google Scholar] [CrossRef]
  18. Schlupp, P.; Blaschke, T.; Kramer, K.; Höltje, H.-D.; Mehnert, W.; Schäfer-Korting, M. Drug Release and Skin Penetration from Solid Lipid Nanoparticles and a Base Cream: A Systematic Approach from a Comparison of Three Glucocorticoids. Ski. Pharmacol. Physiol. 2011, 24, 199–209. [Google Scholar] [CrossRef] [Green Version]
  19. Kovacevic, A.; Savic, S.; Vuleta, G.; Müller, R.; Keck, C. Polyhydroxy surfactants for the formulation of lipid nanoparticles (SLN and NLC): Effects on size, physical stability and particle matrix structure. Int. J. Pharm. 2011, 406, 163–172. [Google Scholar] [CrossRef] [Green Version]
  20. Müller, R.; Runge, S.; Ravelli, V.; Thünemann, A.; Mehnert, W.; Souto, E. Cyclosporine-loaded solid lipid nanoparticles (SLN®): Drug–lipid physicochemical interactions and characterization of drug incorporation. Eur. J. Pharm. Biopharm. 2008, 68, 535–544. [Google Scholar] [CrossRef]
  21. Souto, E.B.; Müller, R.H.; Gohla, S. A novel approach based on lipid nanoparticles (SLN®) for topical delivery of α-lipoic acid. J. Microencapsul. 2005, 22, 581–592. [Google Scholar] [CrossRef] [PubMed]
  22. Carbone, C.; Martins-Gomes, C.; Pepe, V.; Silva, A.M.; Musumeci, T.; Puglisi, G.; Furneri, P.M.; Souto, E.B. Repurposing itraconazole to the benefit of skin cancer treatment: A combined azole-DDAB nanoencapsulation strategy. Colloids Surf. B Biointerfaces 2018, 167, 337–344. [Google Scholar] [CrossRef] [PubMed]
  23. Rigon, R.B.; Gonçalez, M.L.; Severino, P.; Alves, D.A.; Santana, M.H.A.; Souto, E.B.; Chorilli, M. Solid lipid nanoparticles optimized by 22 factorial design for skin administration: Cytotoxicity in NIH3T3 fibroblasts. Colloids Surf. B Biointerfaces 2018, 171, 501–505. [Google Scholar] [CrossRef] [Green Version]
  24. Souto, E.B.; Zielinska, A.; Souto, S.B.; Durazzo, A.; Lucarini, M.; Santini, A.; Silva, A.M.; Atanasov, A.G.; Marques, C.; Andrade, L.N.; et al. (+)-Limonene 1,2-Epoxide-Loaded SLNs: Evaluation of Drug Release, Antioxidant Activity, and Cytotoxicity in an HaCaT Cell Line. Int. J. Mol. Sci. 2020, 21, 1449. [Google Scholar] [CrossRef] [Green Version]
  25. Zielińska, A.; Ferreira, N.R.; Feliczak-Guzik, A.; Nowak, I.; Souto, E.B. Loading, release profile and accelerated stability assessment of monoterpenes-loaded solid lipid nanoparticles (SLN). Pharm. Dev. Technol. 2020, 25, 832–844. [Google Scholar] [CrossRef]
  26. Joshi, M.D.; Müller, R.H. Lipid nanoparticles for parenteral delivery of actives. Eur. J. Pharm. Biopharm. 2009, 71, 161–172. [Google Scholar] [CrossRef]
  27. Mahant, S.; Rao, R.; Souto, E.B.; Nanda, S. Analytical tools and evaluation strategies for nanostructured lipid carrier-based topical delivery systems. Expert Opin. Drug Deliv. 2020, 17, 963–992. [Google Scholar] [CrossRef] [PubMed]
  28. Pardeike, J.; Hommoss, A.; Müller, R.H. Lipid nanoparticles (SLN, NLC) in cosmetic and pharmaceutical dermal products. Int. J. Pharm. 2009, 366, 170–184. [Google Scholar] [CrossRef]
  29. Tan, S.; Billa, N.; Roberts, C.; Burley, J. Surfactant effects on the physical characteristics of Amphotericin B-containing nanostructured lipid carriers. Colloids Surfaces A Physicochem. Eng. Asp. 2010, 372, 73–79. [Google Scholar] [CrossRef]
  30. Han, F.; Li, S.; Yin, R.; Liu, H.; Xu, L. Effect of surfactants on the formation and characterization of a new type of colloidal drug delivery system: Nanostructured lipid carriers. Colloids Surfaces A Physicochem. Eng. Asp. 2008, 315, 210–216. [Google Scholar] [CrossRef]
  31. Fang, J.-Y.; Fang, C.-L.; Liu, C.-H.; Su, Y.-H. Lipid nanoparticles as vehicles for topical psoralen delivery: Solid lipid nanoparticles (SLN) versus nanostructured lipid carriers (NLC). Eur. J. Pharm. Biopharm. 2008, 70, 633–640. [Google Scholar] [CrossRef] [PubMed]
  32. Teeranachaideekul, V.; Souto, E.B.; Junyaprasert, V.B.; Müller, R.H. Cetyl palmitate-based NLC for topical delivery of Coenzyme Q10—Development, physicochemical characterization and in vitro release studies. Eur. J. Pharm. Biopharm. 2007, 67, 141–148. [Google Scholar] [CrossRef] [PubMed]
  33. Doktorovova, S.; Kovacevic, A.B.; Garcia, M.L.; Souto, E.B. Preclinical safety of solid lipid nanoparticles and nanostructured lipid carriers: Current evidence from in vitro and in vivo evaluation. Eur. J. Pharm. Biopharm. 2016, 108, 235–252. [Google Scholar] [CrossRef] [PubMed]
  34. Doktorovova, S.; Souto, E.B.; Silva, A.M. Nanotoxicology applied to solid lipid nanoparticles and nanostructured lipid carriers—A systematic review of in vitro data. Eur. J. Pharm. Biopharm. 2014, 87, 1–18. [Google Scholar] [CrossRef]
  35. Souto, E.B.; Doktorovova, S. Chapter 6—Solid lipid nanoparticle formulations pharmacokinetic and biopharmaceutical aspects in drug delivery. Methods Enzym. 2009, 464, 105–129. [Google Scholar] [CrossRef]
  36. Doktorovova, S.; Souto, E.B. Nanostructured lipid carrier-based hydrogel formulations for drug delivery: A comprehensive review. Expert Opin. Drug Deliv. 2009, 6, 165–176. [Google Scholar] [CrossRef]
  37. Yan, W.; Wistuba, I.I.; Emmert-Buck, M.R.; Erickson, H.S. Squamous Cell Carcinoma—Similarities and Differences among Anatomical Sites. Am. J. Cancer Res. 2011, 1, 275–300. [Google Scholar] [CrossRef]
  38. Okida, F. Estudo da prevalência de casos de cancro da pele e análise da eficácia da protecção solar na prevenção de lesões causadas por radiação ultravioleta em uma amostra da população. An. Bras. Dermatol. 2001, 76, 403–412. [Google Scholar]
  39. Ciążyńska, M.; Narbutt, J.; Woźniacka, A.; Lesiak, A. Trends in basal cell carcinoma incidence rates: A 16-year retrospective study of a population in central Poland. Adv. Dermatol. Allergol. 2018, 35, 47–52. [Google Scholar] [CrossRef]
  40. Balu, M.; Kelly, K.; Zachary, C.B.; Harris, R.M.; Krasieva, T.B.; König, K.; Durkin, A.J.; Tromberg, B.J. Distinguishing between Benign and Malignant Melanocytic Nevi by In Vivo Multiphoton Microscopy. Cancer Res. 2014, 74, 2688–2697. [Google Scholar] [CrossRef] [Green Version]
  41. Metcalf, J.S. Melanoma: Criteria for histological diagnosis and its reporting. Semin. Oncol. 1996, 23, 688–692. [Google Scholar] [PubMed]
  42. Petrella, T.; Quirt, I.; Verma, S.; Haynes, A.E.; Charette, M.; Bak, K. Single-agent interleukin-2 in the treatment of metastatic melanoma: A systematic review. Cancer Treat. Rev. 2007, 33, 484–496. [Google Scholar] [CrossRef]
  43. Wong, H.L.; Bendayan, R.; Rauth, A.M.; Li, Y.; Wu, X.Y. Chemotherapy with anticancer drugs encapsulated in solid lipid nanoparticles. Adv. Drug Deliv. Rev. 2007, 59, 491–504. [Google Scholar] [CrossRef] [PubMed]
  44. Andreani, T.; Dias-Ferreira, J.; Fangueiro, J.; Souza, A.; Kiill, C.; Gremião, M.; García, M.; Silva, A.; Souto, E. Formulating octyl methoxycinnamate in hybrid lipid-silica nanoparticles: An innovative approach for UV skin protection. Heliyon 2020, 6, e03831. [Google Scholar] [CrossRef] [PubMed]
  45. Kumar, C.S.S. Nanomaterials for Cancer Therapy; Wiley-VCH: Weinheim, Germany, 2006. [Google Scholar]
  46. Levy, S.; Furst, K.; Chern, W. A comparison of the skin permeation of three topical 0.5% fluorouracil formulations with that of a 5% formulation. Clin. Ther. 2001, 23, 901–907. [Google Scholar] [CrossRef]
  47. Loven, K.; Stein, L.; Furst, K.; Levy, S. Evaluation of the efficacy and tolerability of 0.5% fluorouracil cream and 5% fluorouracil cream applied to each side of the face in patients with actinic keratosis. Clin. Ther. 2002, 24, 990–1000. [Google Scholar] [CrossRef]
  48. Selvaraj, V.; Alagar, M. Analytical detection and biological assay of antileukemic drug 5-fluorouracil using gold nanoparticles as probe. Int. J. Pharm. 2007, 337, 275–281. [Google Scholar] [CrossRef]
  49. Chatterjee, D.K.; Fong, L.S.; Zhang, Y. Nanoparticles in photodynamic therapy: An emerging paradigma. Adv. Drug Deliv. Rev. 2008, 60, 1627–1637. [Google Scholar] [CrossRef]
  50. Severino, P.; Fangueiro, J.F.; Ferreira, S.V.; Basso, R.; Chaud, M.V.; Santana, M.H.A.; Rosmaninho, A.; Souto, E.B. Nanoemulsions and nanoparticles for non-melanoma skin cancer: Effects of lipid materials. Clin. Transl. Oncol. 2013, 15, 417–424. [Google Scholar] [CrossRef]
  51. Fadeel, D.A.A.; Kamel, R.; Fadel, M. PEGylated lipid nanocarrier for enhancing photodynamic therapy of skin carcinoma using curcumin: In-vitro/in-vivo studies and histopathological examination. Sci. Rep. 2020, 10, 1–10. [Google Scholar] [CrossRef]
  52. Fang, Y.-P.; Tsai, Y.-H.; Wu, P.-C.; Huang, Y.-B. Comparison of 5-aminolevulinic acid-encapsulated liposome versus ethosome for skin delivery for photodynamic therapy. Int. J. Pharm. 2008, 356, 144–152. [Google Scholar] [CrossRef]
  53. Zielińska, A.; Alves, H.; Marques, V.; Durazzo, A.; Lucarini, M.; Alves, T.; Morsink, M.; Willemen, N.; Eder, P.; Chaud, M.; et al. Properties, Extraction Methods, and Delivery Systems for Curcumin as a Natural Source of Beneficial Health Effects. Medicina 2020, 56, 336. [Google Scholar] [CrossRef] [PubMed]
  54. Doktorovova, S.; Souto, E.B.; Silva, A.M. Hansen solubility parameters (HSP) for prescreening formulation of solid lipid nanoparticles (SLN): In vitro testing of curcumin-loaded SLN in MCF-7 and BT-474 cell lines. Pharm. Dev. Technol. 2018, 23, 96–105. [Google Scholar] [CrossRef] [PubMed]
  55. Santos, I.S.; Ponte, B.M.; Boonme, P.; Silva, A.M.; Souto, E.B. Nanoencapsulation of polyphenols for protective effect against colon–rectal cancer. Biotechnol. Adv. 2013, 31, 514–523. [Google Scholar] [CrossRef] [PubMed]
  56. Palliyage, G.H.; Hussein, N.; Mimlitz, M.; Weeder, C.; Alnasser, M.H.A.; Singh, S.; Ekpenyong, A.; Tiwari, A.K.; Chauhan, H. Novel Curcumin-Resveratrol Solid Nanoparticles Synergistically Inhibit Proliferation of Melanoma Cells. Pharm. Res. 2021, 38, 851–871. [Google Scholar] [CrossRef] [PubMed]
  57. Kubler, A.C.; Haase, T.; Staff, C. Photodynamic therapy of primary nonmelanomatous skin tumors of the head and neck. Laser Surg. Med. 1999, 25, 60–68. [Google Scholar] [CrossRef]
  58. Petit, R.; Cano, A.; Ortiz, A.; Espina, M.; Prat, J.; Muñoz, M.; Severino, P.; Souto, E.; García, M.; Pujol, M.; et al. Psoriasis: From Pathogenesis to Pharmacological and Nano-Technological-Based Therapeutics. Int. J. Mol. Sci. 2021, 22, 4983. [Google Scholar] [CrossRef] [PubMed]
  59. Fry, L.; Baker, B.S.; Powles, A.V. Psoriasis—A possible candidate for vaccination. Autoimmun. Rev. 2007, 6, 286–289. [Google Scholar] [CrossRef] [Green Version]
  60. Martins, G.A.; Arruda, L. Tratamento sistêmico da psoríase—Parte I: Metotrexato e acitretina. An. Bras. Dermatol. 2004, 79, 263–278. [Google Scholar] [CrossRef]
  61. Trafford, A.; Parisi, R.; Kontopantelis, E.; Griffiths, C.E.M.; Ashcroft, D. Association of Psoriasis with the Risk of Developing or Dying of Cancer: A Systematic Review and Meta-analysis. JAMA Dermatol. 2019, 155, 1390–1403. [Google Scholar] [CrossRef] [Green Version]
  62. Christophers, E. Psoriasis-epidemiology and clinical spectrum. Clin. Exp. Dermatol. 2001, 26, 314–320. [Google Scholar] [CrossRef] [PubMed]
  63. Nickoloff, B.J.; Schröder, J.M.; Driesch, P.V.D.; Raychaudhuri, S.P.; Farber, E.M.; Boehncke, W.-H.; Morhenn, V.B.; Rosenberg, E.W.; Schön, M.P.; Holick, M.F. Is psoriasis a T-cell disease? Exp. Dermatol. 2000, 9, 359–375. [Google Scholar] [CrossRef]
  64. Prinz, J.C. The role of T cells in psoriasis. J. Eur. Acad. Dermatol. Venereol. 2003, 17, 257–270. [Google Scholar] [CrossRef] [PubMed]
  65. Raychaudhuri, S.P.; Farber, E.M. The prevalence of psoriasis in the world. J. Eur. Acad. Dermatol. Venereol. 2001, 15, 16–17. [Google Scholar] [CrossRef] [PubMed]
  66. Batchelor, J.M.; Ingram, J.R.; Williams, H. Adalimumab vs Methotrexate for the Treatment of Chronic Plaque Psoriasis. Arch. Dermatol. 2009, 145, 704–706. [Google Scholar] [CrossRef]
  67. Menter, A.; Korman, N.J.; Elmets, C.A.; Feldman, S.; Gelfand, J.; Gordon, K.B.; Gottlieb, A.B.; Koo, J.Y.; Lebwohl, M.; Lim, H.W.; et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: Section 4. Guidelines of care for the management and treatment of psoriasis with traditional systemic agents. J. Am. Acad. Dermatol. 2009, 61, 451–485. [Google Scholar] [CrossRef]
  68. Mainardes, R.M.; Urban, M.C.C.; Cinto, P.O.; Chaud, M.V.; Evangelista, R.C.; Gremião, M.P.D. Lipossomes and micro/manoparticles as colloidal carriers for nasal drug delivery. Cur. Drug Deliv. 2006, 3, 275–285. [Google Scholar] [CrossRef]
  69. Grill, A.; Johnston, N.; Sadhukha, T.; Panyam, J. A Review of Select Recent Patents on Novel Nanocarriers. Recent Patents Drug Deliv. Formul. 2009, 3, 137–142. [Google Scholar] [CrossRef]
  70. Venugopal, J.; Prabhakaran, M.; Low, S.; Choon, A.; Deepika, G.; Dev, V.G.; Ramakrishna, S. Continuous Nanostructures for the Controlled Release of Drugs. Curr. Pharm. Des. 2009, 15, 1799–1808. [Google Scholar] [CrossRef]
  71. Trotta, M.; Peira, E.; Carlotti, M.E.; Gallarate, M. Deformable liposomes for dermal administration of methotrexate. Int. J. Pharm. 2004, 270, 119–125. [Google Scholar] [CrossRef]
  72. Wiedersberg, S.; Leopold, C.S.; Guy, R.H. Bioavailability and bioequivalence of topical glucocorticoids. Eur. J. Pharm. Biophar. 2008, 68, 453–466. [Google Scholar] [CrossRef]
  73. Ponec, M.; Kempenaar, J.A.; De Kloet, E.R. Corticoids and Cultured Human Epidermal Keratinocytes: Specific Intracellular Binding and Clinical Efficacy. J. Investig. Dermatol. 1981, 76, 211–214. [Google Scholar] [CrossRef]
  74. Ponec, M.; Polano, M.K. Penetration of various corticosteroids through epidermis in vitro. Arch. Dermatol. Res. 1979, 265, 101–104. [Google Scholar] [CrossRef] [PubMed]
  75. Sivaramakrishnan, R.; Nakamura, C.; Mehnert, W.; Korting, H.; Kramer, K.; Schäfer-Korting, M. Glucocorticoid entrapment into lipid carriers—characterisation by parelectric spectroscopy and influence on dermal uptake. J. Control. Release 2004, 97, 493–502. [Google Scholar] [CrossRef]
  76. Zhang, J.; Smith, E. Percutaneous Permeation of Betamethasone 17-Valerate Incorporated in Lipid Nanoparticles. J. Pharm. Sci. 2011, 100, 896–903. [Google Scholar] [CrossRef]
  77. Yang, J.-H.; Kim, D.-K.; Yun, M.-Y.; Kim, T.-Y.; Shin, S.-C. Transdermal delivery system of triamcinolone acetonide from a gel using phonophoresis. Arch. Pharmacal Res. 2006, 29, 412–417. [Google Scholar] [CrossRef]
  78. Yu, H.-Y.; Liao, H.-M. Triamcinolone permeation from different liposome formulations through rat skin in vitro. Int. J. Pharm. 1996, 127, 1–7. [Google Scholar] [CrossRef]
  79. Zouboulis, C.C. Retinoids—Which Dermatological Indications Will Benefit in the Near Future? Ski. Pharmacol. Physiol. 2001, 14, 303–315. [Google Scholar] [CrossRef] [PubMed]
  80. Brisaert, M.; Gabriëls, M.; Matthijs, V.; Plaizier-Vercammen, J. Liposomes with tretinoin: A physical and chemical evaluation. J. Pharm. Biomed. Anal. 2001, 26, 909–917. [Google Scholar] [CrossRef]
  81. Ioele, G.; Cione, E.; Risoli, A.; Genchi, G.; Ragno, G. Accelerated photostability study of tretinoin and isotretinoin in liposome formulations. Int. J. Pharm. 2005, 293, 251–260. [Google Scholar] [CrossRef]
  82. Malaekeh-Nikouei, B.; Golmohammadzadeh, S.; Hosseini, M.; Nassirli, H. Preparation and characterization of liposomes encapsulated with clindamycin and tretinoin. Pharm. Glob. 2011, 2, 1–4. [Google Scholar]
  83. Latter, G.; Grice, J.E.; Mohammed, Y.; Roberts, M.S.; Benson, H.A.E. Targeted Topical Delivery of Retinoids in the Management of Acne Vulgaris: Current Formulations and Novel Delivery Systems. Pharmaceutics 2019, 11, 490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. 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]
  85. Hadjipavlou-Litina, D.; Magoulas, G.E.; Krokidis, M.; Papaioannou, D. Syntheses and evaluation of the antioxidant activity of acitretin analogs with amide bond(s) in the polyene spacer. Eur. J. Med. Chem. 2010, 45, 298–310. [Google Scholar] [CrossRef] [PubMed]
  86. Hiller, C.; Campbell, C.; Smeaton, I. How stable is dithranol? An investigation into the degradation of different dithranol formulations. Pharm. Pract. 1995, 5, 428–431. [Google Scholar]
  87. Chen, L.; Wang, H.; Zhao, Z.; Zhang, Y.; Huang, G. Effects of the Extract of a Chinese Herb Tripterygium wilfordii Hook f on Rat Pituitary Gland. Am. J. Chin. Med. 2005, 33, 945–955. [Google Scholar] [CrossRef] [Green Version]
  88. Mei, Z.; Wu, Q.; Hu, S.; Lib, X.; Yang, X. Triptolide Loaded Solid Lipid Nanoparticle Hydrogel for Topical Application. Drug Dev. Ind. Pharm. 2005, 31, 161–168. [Google Scholar] [CrossRef]
  89. Katare, O.P.; Raza, K.; Singh, B.; Dogra, S. Novel drug delivery systems in topical treatment of psoriasis: Rigors and vigors. Indian J. Dermatol. Venereol. Leprol. 2010, 76, 612–621. [Google Scholar] [CrossRef]
  90. Spergel, J.M.; Leung, D.Y.M. Safety of topical calcineurin inhibitors in atopic dermatitis: Evaluation of the evidence. Curr. Allergy Asthma Rep. 2006, 6, 270–274. [Google Scholar] [CrossRef]
  91. Mazet, R.; Yaméogo, J.B.G.; Wouessidjewe, D.; Choisnard, L.; Gèze, A. Recent Advances in the Design of Topical Ophthalmic Delivery Systems in the Treatment of Ocular Surface Inflammation and Their Biopharmaceutical Evaluation. Pharmaceutics 2020, 12, 570. [Google Scholar] [CrossRef]
  92. Fang, J.-Y.; Zhang, L.-W.; Al-Suwayeh, S.A.; Hung, C.-F.; Chen, C.-C. Oil components modulate the skin delivery of 5-aminolevulinic acid and its ester prodrug from oil-in-water and water-in-oil nanoemulsions. Int. J. Nanomed. 2011, 6, 693–704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Lin, M.-W.; Huang, Y.-B.; Chen, C.-L.; Wu, P.-C.; Chou, C.-Y.; Wu, P.-C.; Hung, S.-Y. A Formulation Study of 5-Aminolevulinic Encapsulated in DPPC Liposomes in Melanoma Treatment. Int. J. Med Sci. 2016, 13, 483–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Pierre, M.B.R.; Tedesco, A.C.; Marchetti, J.M.; Bentley, M.V.L. Stratum corneum lipids liposomes for the topical delivery of 5-aminolevulinic acid in photodynamic therapy of skin cancer: Preparation and in vitro permeation study. BMC Dermatol. 2001, 1, 5. [Google Scholar] [CrossRef] [Green Version]
  95. Khallaf, R.A.; Salem, H.F.; Abdelbary, A. 5-Fluorouracil shell-enriched solid lipid nanoparticles (SLN) for effective skin carcinoma treatment. Drug Deliv. 2016, 23, 3452–3460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Amasya, G.; Badilli, U.; Aksu, B.; Tarimci, N. Quality by design case study 1: Design of 5-fluorouracil loaded lipid nanoparticles by the W/O/W double emulsion—Solvent evaporation method. Eur. J. Pharm. Sci. 2016, 84, 92–102. [Google Scholar] [CrossRef]
  97. Laue, C.A.-V.; Zoschke, C.; Do, N.; Lehnen, D.; Küchler, S.H.; Mehnert, W.; Blaschke, T.; Kramer, K.; Plendl, J.; Weindl, G.; et al. Improving Topical Non-Melanoma Skin Cancer Treatment: In vitro Efficacy of a Novel Guanosine-Analog Phosphonate. Ski. Pharmacol. Physiol. 2014, 27, 173. [Google Scholar] [CrossRef] [Green Version]
  98. Pukale, S.S.; Mittal, A.; Chitkara, D. Topical Application of Vitamin D3-Loaded Hybrid Nanosystem to Offset Imiquimod-Induced Psoriasis. AAPS PharmSciTech 2021, 22, 1–17. [Google Scholar] [CrossRef]
  99. Alvarez-Figueroa, M.J.; Narváez-Araya, D.; Armijo-Escalona, N.; Carrasco-Flores, E.A.; González-Aramundiz, J.V. Design of Chitosan Nanocapsules with Compritol 888 ATO® for Imiquimod Transdermal Administration. Evaluation of Their Skin Absorption by Raman Microscopy. Pharm. Res. 2020, 37, 1–10. [Google Scholar] [CrossRef]
  100. Amarji, B.; Garg, N.K.; Singh, B.; Katare, O.P. Microemulsions mediated effective delivery of methotrexate hydrogel: More than a tour de force in psoriasis therapeutics. J. Drug Target. 2016, 24, 147–160. [Google Scholar] [CrossRef]
  101. Avasatthi, V.; Pawar, H.; Dora, C.P.; Bansod, P.; Gill, M.S.; Suresh, S. A novel nanogel formulation of methotrexate for topical treatment of psoriasis: Optimization, in vitro and in vivo evaluation. Pharm. Dev. Technol. 2016, 21, 554–562. [Google Scholar] [CrossRef]
  102. 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]
Figure 1. Schematic representation of the encapsulation of active pharmaceutical ingredients (APIs) in different types of lipid nanocarriers [own drawing].
Figure 1. Schematic representation of the encapsulation of active pharmaceutical ingredients (APIs) in different types of lipid nanocarriers [own drawing].
Cancers 13 05619 g001
Figure 2. Different pathways of skin penetration of nanoparticles and superficial spreading melanoma (own drawing).
Figure 2. Different pathways of skin penetration of nanoparticles and superficial spreading melanoma (own drawing).
Cancers 13 05619 g002
Table 1. Types of lipid nanocarriers proposed for the treatment of hyperproliferative skin diseases.
Table 1. Types of lipid nanocarriers proposed for the treatment of hyperproliferative skin diseases.
SystemsDescriptionAdvantagesLimitations
LiposomesVesicles formed by lipid bilayers surrounding an aqueous coreVersatility to load lipophilic, hydrophilic and amphiphilic APIsLow stability and lower loading capacity compared to SLN and NLC
SLNLipid nanoparticles composed of solid lipids with melting point above 40 °CPossibility for scale-up productionRisk of API expulsion from the solid lipid matrix
NLCLipid nanoparticles composed of solid and liquid lipids with melting point above 40 °CHigh loading capacity and encapsulation efficiency for lipophilic drugs; reduced risk of drug expulsion compared to SLNLimited capacity to load hydrophilic APIs
Table 2. Examples of drugs loaded into lipid nanocarriers for the treatment of hyperproliferative diseases.
Table 2. Examples of drugs loaded into lipid nanocarriers for the treatment of hyperproliferative diseases.
DrugType of CarrierMechanism of ActionApplicationReferences
5-aminolevulinic acidOil-in-water emulsionssoybean oil o/w emulsions promoted mALA permeation to deeper skin layersTreatment of subepidermal and subcutaneous lesions[92]
1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC) liposomesReduction of cell viability, mitochondria membrane potential, and enhancement of intracellular ROS accumulationIn melanoma xenograft models, 5-ALA/DPPC enhanced PpIX accumulation only in tumor tissue but not normal skin[93]
Liposomes based on mammalian stratum corneum lipidsDelivery of 5-ALA to viable epidermis and dermisPhotodynamic therapy of skin cancers[94]
5-fluorouracilLecithin and poloxamer 188 based SLNSLN-treated mice exhibited reduced inflammatory reactions, degree of keratosis, and symptoms of angiogenesisskin carcinoma[95]
Tristearin, lecithin, polyvinyl alcohol and Tween 80-based SLNFormulation development by quality by design [96]
Guanosine -analogue phosphonate (OxBu)SLNIncreased caspase-cleaved fragment of keratin-18, caspase-7 activation and reduced expression of matrix metallopeptidase-2 and Ki-67Actinic keratosis, and cutaneous squamous cell carcinoma[97]
ImiquimodVitamin D3-loaded monolithic lipid-polymer hybrid nanoparticlesIn vivo efficacy assessment in imiquimod-induced psoriatic mouse model with enhanced anti-psoriatic activityImroved the Psoriasis Area and Severity Index (PASI)[98]
Chitosan Nanocapsules with Compritol 888 ATOImiquimod-loaded nanocapsules penetration into the stratum corneum and drug reached inner layers of the skinCell carcinoma, ac-tinic keratosis and genital and perianal warts[99]
MethotrexateMicroemulsions in hydrogelsLocation of drug at the desired domain of stratum corneum, epidermal and dermal layers of skin with reduction of systemic absorptionSuccessful treatment of imiquimod-induced psoriatic model, allergic contact dermatitis, rat tail model and safety[100]
Nanostructured lipid carrierReduction of PASI score with recovery of skin miceAmelioration of symptoms of psoriasis imiquimod-induced psoriasis model[101]
siRNAAnti-STAT3 siRNA (siSTAT3) and anti-TNF-alpha siRNA (siTNF-alpha)-loaded in cationic amphiphilic lipid with oleyl chains- based nanoparticlesEfficient delivery of siSTAT3 and siTNF-alpha into the dermisCombination of the two nucleic acids can synergistically treat psoriatic-like plaques[102]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Souto, E.B.; de Souza, A.L.R.; dos Santos, F.K.; Sanchez-Lopez, E.; Cano, A.; Zielińska, A.; Staszewski, R.; Karczewski, J.; Gremião, M.P.D.; Chorilli, M. Lipid Nanocarriers for Hyperproliferative Skin Diseases. Cancers 2021, 13, 5619. https://doi.org/10.3390/cancers13225619

AMA Style

Souto EB, de Souza ALR, dos Santos FK, Sanchez-Lopez E, Cano A, Zielińska A, Staszewski R, Karczewski J, Gremião MPD, Chorilli M. Lipid Nanocarriers for Hyperproliferative Skin Diseases. Cancers. 2021; 13(22):5619. https://doi.org/10.3390/cancers13225619

Chicago/Turabian Style

Souto, Eliana B., Ana L. R. de Souza, Fernanda K. dos Santos, Elena Sanchez-Lopez, Amanda Cano, Aleksandra Zielińska, Rafał Staszewski, Jacek Karczewski, Maria P. D. Gremião, and Marlus Chorilli. 2021. "Lipid Nanocarriers for Hyperproliferative Skin Diseases" Cancers 13, no. 22: 5619. https://doi.org/10.3390/cancers13225619

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