Next Article in Journal
Functional and Safety Profile of Limosilactobacillus vaginalis and Development of Oral Fast-Disintegrating Tablets for Gut Microbiota Modulation
Previous Article in Journal / Special Issue
Generation of Formates Following 20 kHz Sonication of DSPE-mPEG2000 PEGylated Phospholipid Micelles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Local Chemotherapy of Skin Pre-Neoplastic Lesions and Malignancies from the Perspective of Current Pharmaceutics

Department of Pharmaceutical Technologies, Faculty of Pharmacy, Medical University of Varna, 9000 Varna, Bulgaria
Pharmaceutics 2025, 17(8), 1009; https://doi.org/10.3390/pharmaceutics17081009 (registering DOI)
Submission received: 30 June 2025 / Revised: 27 July 2025 / Accepted: 29 July 2025 / Published: 1 August 2025

Abstract

In the preceding and early stages of cancer progression, local drug delivery to pre-cancerous and cancerous skin lesions may be applied as an alternative or supplementary therapy. At present, 5-Fluorouracil, imiquimod, and tirbanibulin creams and ointments have established their place in practice, while several other active pharmaceutical ingredients (APIs) (e.g., calcipotriol, tretinoin, diclofenac) have been repurposed, used off-label, or are currently being investigated in mono- or combined chemotherapies of skin cancers. Apart from them, dozens to hundreds of therapeutics of natural and synthetic origin are proven to possess anti-tumor activity against melanoma, squamous cell carcinoma (SCC), and other skin cancer types in in vitro studies. Their clinical introduction is most often limited by low skin permeability, challenged targeted drug delivery, insufficient chemical stability, non-selective cytotoxicity, or insufficient safety data. A variety of prodrug and nanotechnological approaches, including vesicular systems, micro- and nanoemulsions, solid lipid nanoparticles, nanostructured lipid carriers, polymeric nanoparticles, and others, offer versatile solutions for overcoming the biophysical barrier function of the skin and the undesirable physicochemical nature of some drug molecules. This review aims to present the most significant aspects and latest achievements on the subject.

1. Introduction

Skin cancers are generally classified into melanoma and non-melanoma types. Altogether, they represent the most commonly encountered form of malignancy worldwide, and their incidence increases with the rise in the elderly population [1,2]. Melanoma is among the fastest progressing cancers overall; it accounts for approximately 1% of all skin cancer cases and is distinguished by a high mortality rate after metastasis [3,4,5]. Non-melanoma skin cancers (NMSCs), contrarily, are mostly slow-developing, locally invasive, and curable. The basal and squamous cell carcinomas (BCC and SCC, respectively) represent the prevailing 99% of all NMSCs, while the rest include some less common but aggressive skin malignancies, such as cutaneous lymphomas, Kaposi’s sarcoma, Merkel cell carcinoma, skin carcinosarcoma, and dermatofibrosarcoma [6,7,8].
Skin cancer etiology is considered to have a tight bond with the harmful impact of ultraviolet (UV) light from sun exposure. While the UV-C rays are short enough (λ = 200–290 nm) to be entirely absorbed by the ozone layer, the longer UV-B (λ = 290–320 nm) and UV-A (λ = 320–400 nm) rays manage to penetrate the troposphere and reach the human epidermal and dermal skin layers, respectively. Thereby, they induce cascade reactions of reactive species generation (e.g., reactive oxygen species—ROS; reactive nitrogen species—RNS), DNA oxidation and fragmentation, mutations, and subsequent cell damage [8,9]. In some cases, depending on the oxidative status and immunity of the body, these processes may lead to abnormal skin morphology such as keratinocytes deformation, hypertrophy (e.g., of stratum corneum or stratum spinossum), sebaceous hyperplasia, reduction in the Langerhans cells’ count and suppression of their function, dryness, hyperpigmentation, and not exclusively, benign or malignant neoplasia (skin tumors) [10,11,12]. Additional factors involved in skin cancer etiology are ionizing radiation, exposure to some chemicals (e.g., arsenic), viral infections (e.g., human papillomavirus—HPV), or other environmental triggers. Important pre-cancerous skin conditions to be timely addressed and treated are actinic keratosis (AK) and Bowen’s disease (BD), which manifest as dry scaly patches or red scaly patches, respectively, and hold the risk of progression into SCC [13,14] (Figure 1).
The cutaneous and transcutaneous dosage forms offer several advantages, among which are an easy and non-invasive application, avoidance of hepatic metabolism upon first pass and some systemic side effects, targeted therapy, possibility for controlled drug flux, and dose reduction. However, very few compounds are able to cross the SC, reach targets in the skin, and act selectively and safely without an explicit chemical modification, physical enhancement of diffusion, or technological formulation. There lies the difference between the numerous in vitro active anti-cancer drug molecules and the very few ones finding a practical application and reaching the clinic [15]. In this regard, the role of nanotechnologies for topical skin cancer prevention and treatment in the early or post-operative stages narrows down to ensuring the desired qualities of the therapeutic entity for dermal administration [16,17]. The efficient topical drug therapy of skin lesions requires a multi-target approach aiming at selective cytotoxicity, inflammation, and infection [18,19,20]. From a technological point of view, enhanced skin permeation, sustainable drug liberation, and stability are to be sought after. The various nanotechnologies may ensure or improve one or more aspects of the above [21,22,23,24,25]; the strong and weak sides of each particular nano-carrier will be reviewed, including the lipid-based nanoparticles and systems, the polymeric, and inorganic nanomaterials.

2. Skin Anatomy, Histology, and Physiology in Brief

The skin, being the largest organ in the human body, possesses a surface of about 2 m2 in adults and a thickness varying in the range of 0.5–4 mm. Well-known, inside out, it comprises the subcutaneous layer (also referred to as subcutis or hypodermis), the dermis, and the epidermis. The epidermis, as the outermost, non-vascularized layer of the skin, exerts a protective function against loss of water and electrolytes, physical and chemical stimuli, and entry of foreign bodies, including microorganisms. It consists of the superficial, non-vital, horny layer—stratum corneum (SC)—and the underlying vital epidermal tissue. The latter is additionally subdivided into four layers—the stratum basale, spinosum, granulosum, and lucidum, respectively [26,27,28,29]. The main cells in the epidermis are the keratinocytes. They undergo division in the basal layer, and one of two daughter cells migrates to the surface; during this process, the keratinocytes acquire a squamous morphology and finally differentiate into corneocytes—dead, anucleated cells filled with keratin. The tightly ordered corneocytes in a semi-fluid lipid matrix of ceramides, fatty acids, and cholesterol build the structure of SC, the nature of which will be further reviewed with respect to drug transport. Another major epidermal cell type is the melanocyte, responsible for the synthesis of melanin. The Langerhans cells and the Merkel cells in the epidermis are involved in the immune and sensory responses, respectively [26,27,28,29].
The primary function of the dermis, a connective tissue layer underlying the epidermis, is secretory and thermoregulatory. It also contributes to the skin’s defense mechanisms against pathogenic microorganisms by maintaining a slightly acidic pH of 5.4–5.9 on the surface as a result of the sweat and sebaceous glands’ secretion. The pH increases in the vital cutaneous layers up to 7.1–7.3 [30,31]. The hypodermis, a vascularized and neuro-supplied fatty tissue, serves as a fat depot and nourishes the upper skin layers. It regulates the body temperature through a neuro-reflective pathway in which the thermoregulatory center controlling the sweat gland secretion is involved [26,32].

3. Dermal and Transdermal Drug Transport

As for all environmental pests of chemical, biological, or physical origin, the main barrier for drug penetration into the deeper skin layers is SC. In between the corneocytes, the extracellular space in SC is organized in lamellar bilayers comprising cholesterol, fatty acids, ceramides, and structural proteins (filaggrin, loricrin, and involucrin). The structural integrity and the cohesiveness of the cellular and extracellular elements that build SC define its resilience and barrier function. A key role in this regard is attributed to the epidermal lamellar bodies situated in the stratum granulosum’s keratinocytes and responsible for the cornified layer’s lipid nourishment. Under standard physiological conditions, SC is partially or entirely impermeable to most substances, including drug molecules [33,34,35,36].
The most likely drug transport mechanisms across the SC are of the transepidermal type and involve bypassing diffusion through the extracellular lipid milieu (paracellular route) or transcellular passage. While the paracellular route is favored by drug molecules with moderate to pronounced lipophilicity (log P ≥ 2) and relatively low molecular weight (<500 Da), the trans-corneocyte pathway is an alternative transit for hydrophilic drugs whose nature does not allow sufficient solubility and diffusion through the lipid matrix. The latter transport mechanism requires the keratin hydration that regulates the permeability of corneocytes [37,38,39]. Supporting the dermal drug delivery are the transappendageal routes, which include diffusion through the hair follicles or the sweat glands [40]. Although considered a secondary mechanism of drug transportation, the transfollicular path has gained much attention in recent years. Particular reasons for that are the eloquent findings of increased drug uptake from hair-rich skin regions as compared to hairless skin areas, as well as the nanotechnological advancements that allow transfollicular drug targeting. The transfollicular drug transport is desirable when treating skin diseases affecting the pilosebaceous units (e.g., alopecia and acne) or when aiming at transcutaneous drug absorption [41,42,43,44].
Regardless of the transportation mechanism, three stages of drug entry through the skin layers could be distinguished. Penetration occurs when the drug has reached the SC and the vital epidermis (epidermal transport); permeation is defined as the transit of the drug to the vascularized layers (dermal transport); percutaneous absorption (transdermal; transcutaneous transport) is the process of the drug entering the bloodstream [45]. Each disease requires a drug delivery and preferential deposition targeted at specific skin structures. In the cases of pre-cancerous and cancerous skin lesions, such targets are the abnormal cell formations and the margins thereof. Up until stage II skin carcinomas, the abnormal tissue may spread from the epidermal layers to the dermis [29]. The affected skin areas are normally characterized by increased permeability [34].

4. Biopharmaceutical Aspects of Dermal and Transdermal Drug Delivery

4.1. Physiological Aspects

The structure and function of the skin dynamically change with age, as well as a result of environmental factors, care with cosmetic products, concomitant diseases, and intake of certain medications. Consequently, there are considerable intra- and interindividual variations in skin permeability. As the skin matures, its barrier function gradually improves—the water content reduces, the fat secretion enhances, and the immune defense develops. With aging, epidermal atrophy, reduced secretion of the sweat and sebaceous glands, and impaired superficial lipid mantle become notable; a decrease in the capillary resistance of the dermis and a loss of elasticity tend to occur [46,47]. These peculiarities of elderly skin, which is indeed the most affected by pre-cancerous and cancerous neoplasia, impose the use of predominantly lipid-based or amphiphilic drug vehicles or lipid-based nanotechnologies. In cases of dry skin (xerosis) or dry scaly skin patches (e.g., AK, BD, SCC), the same recommendations apply [48,49]. In oily skin regions or in cases of exuding or ulcerating skin lesions, primarily hydrophilic or amphiphilic vehicles and nano-carriers might be preferred for better results [50].
Although the water content in SC under standard conditions is low (10–15%), this layer is highly hygroscopic and can absorb water up to 50–75% of its own weight (after a bath or occlusive therapy, for example). The hydrated SC is characterized by increased permeability and a limited protective capacity. The underlying vital skin layers are substantially more hydrophilic, and their water content reaches up to 70% [51,52]. It should be acknowledged that the degree of skin hydration varies in a wide range depending on the air humidity, the outside temperature, the use of skin care products, and, not least, concomitant diseases and the intake of particular medicines [53,54,55]. Often, a pronounced skin dryness is observed in patients with diabetes, hypothyroidism, imbalanced sex hormones, or those receiving diuretics and retinoids. The administration of peripheral vasodilators and anticoagulants may potentiate drug absorption through the skin and, in cases of local dermal therapy, increase the risk of systemic side effects [56,57,58,59,60,61,62,63].
Skin metabolism is an additional defense mechanism against potentially hazardous molecules that have managed to bypass the SC. Isoforms of alcohol dehydrogenase, aldehyde dehydrogenase, flavin-dependent monooxygenases, carboxylesterases, and the cytochrome enzymes (CYP 2D6, 2E1, 3A4, 1A1, 1A2, 2C9) are expressed in the skin layers and/or the skin appendages. The enzymatic activity of the skin depends on the age (the activity of esterases, for example, increases as the body matures), the anatomical site (some enzymes are found only in specific skin areas), and some environmental factors (e.g., exposure to UV radiation). Cutaneous metabolism plays a key role in the prodrugs’ biotransformation into active metabolites. A classic example in this regard is corticosteroids [64].

4.2. Pathophysiological Aspects: Pre-Cancer- and Cancer-Related Impairment of the Skin’s Barrier Function

Several particularities of the pre-cancerous and cancerous skin lesions usually determine an impaired structural integrity and enhanced permeability compared to healthy skin. The pathologically increased vascularity is among the leading factors explaining the enhanced permeability and retention effect (EPR) inherent for most tumors and allowing a tumor-specific targeting of macromolecules and drug-carrying nanoparticles [65,66]. Other often encountered structural changes in the concerned skin regions include defective lamellar bilayers of the cornified layer’s extracellular lipid matrix, hyperplasia, hypergranulosis, hyperkeratosis, increased transepidermal water loss, and decreased hydration; a scaly appearance and increased pH on the surface are typical for most skin lesions [34].

4.3. Drug Aspects in Dermal Drug Delivery

Skin penetrability is predetermined by the drug’s chemical nature, and the most important characteristics related to it are the molecular weight, the hydrophilic–lipophilic balance, and the ionizability. The particle size of a substance when in a powder or suspension-type of dosage form and polymorphism, where applicable, stand out as factors as well. It is widely considered that passive diffusion through SC is possible for molecules below 500 Da, if not less, when other factors play a negative role. The octanol/water partition coefficient value (LogPo/w), on the other hand, being in direct relation with the practically significant distribution between dosage form and SC, should ideally fall in the range of 1–4 [67,68]. However, the lesser the polarity of a molecule and the greater the LogPo/w value, the stronger the retention in SC and the more limited further drug diffusion to the vital skin layers is [69,70]. The presence of ionizable functional groups in the structure of the drug may hinder skin penetrability depending on the respective dissociation constant/s value/s (pKa). When charged, a molecule is more likely to step into intermolecular interactions, causing steric immobilization and preventing passive diffusion [71]. Ionization and the hydrophilic–lipophilic properties of an active pharmaceutical ingredient (API), as well as other undesirable physicochemical properties, could be modulated to a certain extent by obtaining prodrug derivatives [72,73,74]. Esterification or etherification reactions are frequently applied to engage hydroxyl (-OH/O-) and carboxyl (-COOH/COO-) functional groups with the purpose of achieving an enhanced permeation rate, reduced drug crystallinity, etc. [75,76]. A few examples of prodrugs for the topical treatment of skin pre-cancerous and pre-cancerous lesions are listed in Table 1.

4.4. Dermal Dosage Forms and Excipients

Dermal dosage forms could generally be classified into five categories, including liquid formulations (lotions, shampoos, foams), semi-solid forms (ointments, creams, gels, and others), powders for cutaneous application, irrigation solutions, and transdermal patches [89]. The last three, being either superficially active or designed for systemic drug uptake via percutaneous absorption, are not suitable formulations for targeting cancerous and pre-cancerous lesions. The topically active anti-cancer drugs rely mostly on liquid or semi-solid vehicles to ensure their stability during storage, retention on skin, liberation, and facilitated permeation [90,91]. Bases with amphiphilic properties, such as liquid emulsions, creams, bigels, nanobigels, emulgels, and foams, resemble the skin’s nature the best and are considered most appropriate for drug delivery to the vital epidermis and the dermis. Moreover, the utilization of lipid-containing multi-phase vehicles of the various types (oil-in-water, water-in-oil, or multiple emulsion bases) is in good agreement with the physiological needs of xerosis skin, inherent for most pre-cancerous or cancerous lesions. Exceptions include lesions accompanied by ulceration and open wound formation, for which the use of water-based formulations, such as sterile solutions and hydrogels, is recommended. In all cases, bases of low-to-moderate viscosity are preferable when one or more of the following is valid: a need for faster permeation and onset of action; application on larger and richly haired skin areas; application on massive, rough, uneven, and sensitive lesions [89,90,91]. Major drawbacks of the conventional dermal dosage forms in the context of skin cancer therapy are the limited potential for modified drug release, drug targeting, stabilization, and enhanced permeation rate [89,90,91]. Moreover, many of them suffer from low patient compliance and a high incidence of local adverse reactions [92]. Alternatively, dermal or microneedle-equipped intradermal patches (films) are being developed and studied as potential platforms for the topical delivery of APIs, including antineoplastic agents, for they allow prolonged retention and extended therapeutic effects [92,93]; however, in their classical variations, these dosage forms do not offer a solution for better skin tolerability or active drug targeting.
An important class of pharmaceutical excipients used in dermal dosage forms is the permeation enhancers. They represent a heterogeneous group of adjuvants united by their ability to facilitate the drug’s passive diffusion through SC [26,94,95]. Classic permeation enhancers are dimethylsulfoxide (DMSO) and other similar solvents—e.g., dimethylformamide, diethylene glycol monoethyl ether (ethoxydiglycol; Transcutol®), dimethyacetamide, and others; being very “good” solvents for a vast spectrum of molecules, they increase the skin’s permeability by dissolving and extracting lipids from the cornified layer and creating hydrophilic “channels”. DMSO, additionally, is known to denature keratin and other proteins in SC [96,97]. A disadvantage is the high active concentration of this permeation enhancer (>60%), which often causes skin irritation (erythema, skin dryness, urticaria, etc.) and limits its use [94]. Azone® (1-dodecylazacycloheptan; laurocapram) is a strongly lipophilic compound (LogPo/w 6.2) able to integrate into the phospholipid bilayer of corneocytes and increase the membranes’ fluidity; it is an effective absorption enhancer for both lipophilic and hydrophilic drugs and exerts its activity in relatively low concentrations (1–3%) [98,99]. Similar mechanisms of action are intrinsic for other permeation enhancers, such as oleic acid, pyrrolidones, terpenes, and terpenoids. The latter two main components of the essential oils may also increase the lipids’ solubility or cause vasodilation [100]. Other commonly used permeation enhancers are oxazolidinones, some keratolytics (urea, salicylic acid, etc.), and various humectants [101]. Except for chemical enhancers, physical processes and mechanistic approaches may also be applied to enhance absorption into and through the skin. Examples are ultrasound-assisted, iontophoresis-assisted, electroporation-assisted, and microneedle-assisted (trans)dermal drug delivery.

5. Nanotechnologies for Dermal Drug Delivery

Advancing technologies allow many of the desired qualities of a dermal formulation, namely, drug stability, prolonged retention on skin, extended drug release, enhanced permeation, targeted delivery, lowered toxicity, and improved tolerability, to be achieved by the use of nano-scaled drug delivery systems (Figure 2) [69,102]. Accordingly, the most recent reports on the local chemotherapy of skin pre-neoplastic lesions and malignancies involve nanotechnology-assisted approaches. In the context of dermal drug application, the suitable nano-carrier systems include vesicular carriers of lipid, polymeric, or hybrid nature, other lipid-based nanoparticles (e.g., solid lipid nanoparticles, nanostructured lipid carriers, nano- and microemulsions), dendrimers, and metal nanoparticles [5,103,104].

5.1. Vesicular Drug Carriers

The era of vesicular drug carriers began with liposomes in the 1960s. By being uni- or multi-lamellar vesicles comprising lipid bilayer(s) surrounding an aqueous milieu, they are able to accommodate lipophilic or hydrophilic APIs. Among their strengths are biocompatibility, biodegradability, and low immunogenicity. Following several generations of development, liposomal technologies still evolve and manage to overcome significant drawbacks from the past, such as the short biological half-life and the limited potential for targeted drug delivery. Thereby appear the cationic, stimuli-responsive, actively targeted, and long-lasting (stealth) liposomes [105]. Still, challenges to the large-scale and clinical introduction of liposomes remain: the long-term stability, the quality assurance of the manufacturing process for more complex (i.e., modified) formulations, the low drug loading and encapsulation capacity, and the production cost [106]. Meanwhile, different courses of modification have led to the establishment of other classes of vesicular systems with improved abilities in dermal and transdermal drug delivery. These include the ethosomes, transferosomes, invasomes, niosomes, polymersomes, pharmacosomes, phytosomes, cubosomes, glycerosomes, and chitosomes. Most of them share similar drug permeation enhancement mechanisms, involving one or more of the following: superficial skin deposition and drug release; diffusion of intact flexible vesicles through the skin layers and subsequent drug release; transfollicular targeting; vesicles’ fusion into cell membranes and subsequent cytoplasmic drug release; endocytosis by phagocytic cells [107,108]. In more detail, information about the various vesicular nano-carriers is revealed in Table 2.

5.2. Other Lipid-Based Nanotechnologies

Other lipid-based nanotechnologies emerging in the field of dermal and transdermal drug delivery, apart from the vesicular systems, are solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs). Chronologically, SLNs appeared first in the 1990s as a result of Professor R.H. Müller and Professor M. Gasco’s attempt to overcome the drawbacks of liposomes and polymeric nano-carriers [108]. SNLs are core–shell organized nanoparticles composed of a solid (at room and body temperature) lipid nucleus and a surfactant crown. NLCs are a later development using liquid lipids in the particles’ core as well. The less organized (amorphous) and fluid state of the lipid matrix thereby obtained improves the drug loading capacity of the lipid nanoparticles. Both the SLNs and the NLCs are currently widely investigated in dermal drug delivery because of several key advantages they offer over liposomes, viz. improved drug stability (by SLNs), better physicochemical stability of the system (by SLNs and NLCs), additional occlusive-determined drug permeation enhancement mechanism through skin (by SLNs and NLCs), additional stratum corneum and cell membrane fluidization for improved skin penetrability (by NLCs), and greater potency in prolonged drug delivery (by SLNs and NLCs) [134,135,136,137].
The nano- and microemulsions, unlike classic emulsions, are also regarded as lipid-based nanotechnologies. While they cannot offer as many perspectives in drug release modification, active targeting, and drug stabilization as compared to the other members in the group, they still offer a few benefits as dermal delivery platforms. Both nano- and microemulsions do not require further formulation into a dosage form (e.g., creams, gels, lotions, foams, patches, etc.) as they serve as liquid dermal vehicles as well. The oil-in-water emulsions exert an additional hydration effect on the intracellular keratin and solubilize the superficial sebum, while the water-in-oil emulsions fluidize the extracellular lipid matrix of SC, which either way leads to an improved epidermal permeability [108,138,139]. Furthermore, nano- and microemulsions are obtained via comparatively simpler technologies and are distinguished by a narrow size distribution, highly increased active surface area, and uniform spreading on the skin [140,141]. Lastly, the microemulsions, which are formed spontaneously with significantly higher surfactant concentrations as compared to nanoemulsions, represent thermodynamically stable systems [142].

5.3. Polymeric Nanoparticles

Polymeric materials, such as natural or semi-synthetic polysaccharides, synthetic acrylates, and others, find broad application as structural units of dermal drug delivery dosage forms—e.g., semi-solid hydrogels and dermal and transdermal patches [143,144,145]. However, the majority of polymeric nanomaterials are usually not common in dermal or transdermal drug delivery because of their limited ability to permeate the skin layers [146,147]. Interesting representatives worth mentioning in the context of this review are dendrimers. These three-dimensionally branched macromolecules exert several unique properties that could be useful in targeted local cancer therapy. Particularly, they possess high surface functionality eligible for modification and the ability to reduce the toxicity of chemotherapeutic agents, to improve drug solubility, and to ensure controlled drug release [148,149,150]. Dendrimers are also known to improve the delivery and efficacy of photosensitizers used in PDT for cancer treatment and act as permeation enhancers [151,152]. Moreover, unlike the majority of polymeric nanotechnologies that suffer from low reproducibility, challenging scalability, and toxicity problems, dendrimers result in a highly monodisperse system with a precisely defined size range and are known for their good biocompatibility [153,154,155].

5.4. Inorganic Nanoparticles

Inorganic nanomaterials are a diverse group of nanoparticles that includes metal nanoparticles, mesoporous silica nanospheres, magnetic nanoparticles, quantum dots, graphene oxide nanosheets, and others [156,157]. Still, two things that unite the majority of members of this class are their intrinsic pharmacological activity and responsiveness to external stimulation (e.g., magnetic field, electric field, light, temperature, pH), with the latter being a valuable tool in active drug targeting and controlled release [158,159]. Because of all these, the applications of inorganic nanomaterials progressively extend from imaging and diagnostics to theranostics and gene and drug delivery [160,161]. However, it should be noted that the most substantial toxicity concerns are reported for the representatives of this class owing to decomposition, nuclear penetration, or impurities [155,162].
Metal nanoparticles, especially those comprising noble metals—silver, gold, palladium, and platinum—are of particular interest in skin pathologies. Although the relatively high toxicity of these materials often limits their use to external administration, they have been systemically reported to possess explicit antimicrobial and anti-tumor properties [163,164,165,166,167,168]. Furthermore, their large and highly reactive surface area allows functionalization with other therapeutic agents as well [168,169,170]. A common event in this respect is the utilization of silver and gold nanoparticles (AgNPs; AuNPs) in wound healing [171,172,173]. Some key features of AgNPs and AuNPs are the ability to generate ROS and improve the efficacy of PDT, as well as the ability to disrupt membranes and facilitate drug permeation and action [174,175,176]. On the other side, metal oxide-based nanomaterials, such as titanium dioxide (TiO2) and zinc oxide (ZnO), are an object of interest in sunscreens and other products for skin cancer prevention, although there have been serious safety concerns related to their use [177,178,179,180,181,182]. For example, the European Commission banned TiO2 (also known as E171) as a food additive in 2022 due to potential genotoxicity. While it is still allowed in non-food products, the search for safer alternatives is being prioritized [183].

6. Local Chemotherapy of Skin Pre-Neoplastic Lesions and Malignancies

6.1. 5-Fluorouracil (5-FU)

5-FU, an antimetabolite that stands in the way of thymidine synthesis and DNA replication, and one of the oldest broad-spectrum anti-cancer drugs, is considered a gold standard API in the local chemotherapy of skin pre-neoplastic lesions and cancers [85,184,185]. Topical 5-FU products, e.g., Carac®, Efudex®, and Fluoroplex®, range in 0.5% to 5% active concentration and are officially approved for the treatment of AK and superficial BCC [85,186,187]. However, numerous studies have revealed the efficacy of 5-FU against other skin cancer types as well, including SCC and melanoma [188,189,190,191,192,193,194,195]. Furthermore, data have become evident to support the mechanisms of action of the compound. For example, Peng et al. showed that 5-FU nanoparticles regulate the Wnt/β-catenin signaling pathway and thereby inhibit the proliferation of SCC cells and induce apoptosis [196]; Tian et al. found that 5-FU triggers anti-tumor immunity by activating the Stimulator of Interferon Genes (STING) pathway in cancer cells and enhancing the immune response against melanoma cancer cells [197].
5-FU is a BCS class III drug, and key disadvantages of the conventional topical therapy arise from its low skin penetrability. In this respect, 5-FU prodrugs, combinations with other therapeutics, and formulation into advanced drug delivery systems, including nano-scaled drug delivery systems, represent a tremendous interest today [198,199]. According to the PubMed database, only the count of clinical trials for the past 10 years involving 5-FU in skin cancer therapy exceeds 25 [200]. Apart from that, dozens to hundreds of scientific articles concern the subject annually. Table 3 summarizes the outcomes from some recent research studies on nanotechnology-assisted topical 5-FU delivery.

6.2. Imiquimod (IMQ)

IMQ is a topically active immunomodulator, particularly effective against superficial BCC. Imiquimod formulations for cutaneous use on the market (e.g., AldaraTM 5% cream and Imiquimod 5% cream) are also indicated for the treatment of AK and genital warts. Lower active drug concentrations of 3.75% are recommended when the facial skin is to be treated [223,224]. IMQ’s primary mechanism of action is grounded on the interaction with Toll-like receptors 7 and 8, toward which the compound acts as an agonist. Thereby, it upregulates the inflammatory response of the immune cells (e.g., neutrophils, macrophages, dendritic cells) and induces apoptosis in tumor cells [223,224,225,226]. The so-far established pharmacodynamics of IMQ have encouraged scientists and physicians to suggest and prove the efficacy of the compound against other forms of skin cancer besides BCC; particularly, there have been promising results published on IMQ’s effectiveness in cases of nodular BCC and SCC and persistently positive margins of melanoma in situ [227,228,229,230,231].
Chemically, IMQ is characterized by a low-molecular-weight (240.3 g/mol) imidazoquinoline structure. Despite that, the API suffers an extremely low skin penetrability (<3%) [232]; this is likely due to its strict lipophilic nature, insufficient water solubility (reported to be between 0.6 and 7.5 µg/mL in the temperature range between 4 and 30 °C, respectively) [233], and unfavorably slow partitioning into the vital epidermis as a result of a high affinity to the commercially used lipophilic cream bases or the superficial SC upon application [234,235,236]. Furthermore, as a base (pKa 7.3), IMQ steps into an ionized state in an acidic environment, which may also contribute to its negligible permeation rate [237,238]. From the point of view of the safe local therapy of superficial BCC, the so-minimized risk of systemic uptake via the transdermal route and the subsequent occurrence of systemic adverse reactions might be beneficial [232]. However, it limits the therapeutic potential of the drug in nodular skin cancer formations and invasive skin tumors [235]. Table 4 presents a few recent attempts to improve the skin permeation rate and the local bioavailability of IMQ by using nanotechnological instruments.

6.3. Tirbanibulin

Tirbanibulin is a novel synthetic drug, recently approved (in 2020 by the FDA and in 2021 by the EMA) for the topical treatment of AK, particularly in the face and scalp area. It is available as 1% ointment under the brand name Klisyri® [249]. Tirbanubulin has shown promising anti-tumor and anti-proliferative potential against skin cancer cell lines, including melanoma and SCC. Its primary mechanism of action is known to be microtubulin polymerization inhibition and cell cycle arrest in rapidly dividing cells. Additionally, studies have revealed interference of the API with the proto-oncogenic Src tyrosine kinase signaling pathway, p53 upregulation, and induced apoptosis via caspase-3 stimulation and poly (ADP-ribose) polymerase cleavage [249,250,251]. Clinical trials have concluded a relatively good tolerability of the drug and a lower occurrence of local adverse reactions in comparison to a control ointment. A key reason for the latter observations appears to be the substantially shortened period of treatment (up to 5 days) with tirbanibulin 1% in comparison to a two- to several-week duration of the treatment by other products with the same indication [249,250,251]. Currently, the API tirbanibulin and the product Klisyri® are under patent protection.

6.4. Diclofenac

Diclofenac, a non-steroidal anti-inflammatory drug, is conventionally used to manage pain and inflammation in musculoskeletal disorders [252]. As a COX-2 inhibitor, the API blocks the synthesis of prostaglandin E2 (PGE2) (a hormone-like substance recognized as a promotor of tumor growth), which explains the established beneficial effects of the topical diclofenac therapy in the treatment of skin pre-cancerous lesions and tumors characterized by overexpression of the COX-2 enzyme and elevated levels of PGE2 [253,254]. The anti-tumor effects of the drug are also supported by data showing the activation of mitochondrial apoptosis pathways through the modulation of Bcl-2 protein expression [255]. To date, the topical application of diclofenac 3% has been proven to prevent the progression of AK into SCC and to reduce tumor burden in SCC and superficial BCC lesions [255,256]. Diclofenac 3% gel in 2.5% hyaluronic acid (under the trade name Solaraze® 3% Gel) is FDA-approved for the treatment of AK [257]. Topical diclofenac monotherapy has shown limited efficacy in nodular and invasive skin tumors, whereas several combinations have been suggested for improved results—e.g., topical diclofenac and calcipotriol, topical diclofenac plus PTD, topical diclofenac plus CO2 laser ablation, and IMQ 5% [258,259,260]. A hybrid compound of diclofenac and hydroxytyrosol, a natural antioxidant and potential anti-cancer agent, synthesized by Tampucci et al., has shown enhanced cytotoxicity against SCC cell lines as compared to native diclofenac. Furthermore, introduced in the form of self-assembling nanomicelles, the newly discovered diclofenac ester demonstrated a reduced risk of systemic transcutaneous absorption and increased deposition in skin layers in comparison to Solaraze® 3% gel [261]. While many other scientific reports on diclofenac nano-formulation exist, they do not specifically regard topical skin cancer treatment as a potential therapeutic indication.

6.5. Ingenol Mebutate (IM)

IM is a relatively new therapeutic agent that was approved by the FDA in 2012 for the topical treatment of AK. It emerged on the market as Picato® 0.05% and 0.015% gel, with the lower concentration being indicated for sensitive skin zones (face and scalp). A key advantage of the local treatment with IM appeared to be the fast therapeutic response and the short duration of the therapeutic course (2–3 days) [262,263]. It is known that IM triggers protein kinase C- and neutrophil-mediated immune responses leading to rapid cell necrosis and cellular cytotoxicity [262,263,264,265]. Except for AK, IM has shown promise in the treatment of BD, BCC, and SCC [266,267,268,269,270,271]. However, recent reports have associated the use of IM with an increased risk of SCC in AK patients [272,273].
IM is a tetracyclic diterpene first isolated from Euphorbia peplus (milkweed; cancer weed; petty spurge)—a traditional herbal medicine, particularly used to treat pathological skin conditions [263,274]. Several successful attempts have been made for the chemical synthesis of IM and its further derivatization, aiming at a more efficient and scalable method for production and improved chemical stability with comparable pharmacological activity [275,276]. The compound possesses an eligible molecular weight for dermal delivery (430.5 Da) and overcomes the skin barrier via P-glycoprotein-mediated transport but is chemically unstable in alkaline media and is characterized by a very limited water solubility [262]. It is believed that the need for an organic vehicle (particularly isopropanol) for its formulation into a gel, as well as the low pH of the composition, maintained to improve stability, are among the leading causes of adverse skin reactions accompanying the topical use of IM [263]. By chemical modifications and investigation of the pharmacological properties and stability, Liang et al. have identified some key features for potent and more stable ingenol derivatives [277]. Bertelsen et al. have successfully synthesized a novel ingenol derivative, namely ingenol disoxate, which exhibits superior chemical stability, augmented potency, and comparable biological activity in relation to IM [278].

6.6. Vitamin D3 and Analogs

Vitamin D3 analogs (VD3A), such as calcipotriol (calcipotriene), calcitriol, tacalcitol, and maxacalcitol, are a first-line treatment for psoriasis because they are known to modulate the vitamin D receptor (VDR)-mediated responses, thereby promoting cell differentiation, inhibiting proliferation, enhancing apoptotic processes, and exerting immunomodulatory activity [279,280]. This mechanism of action has prompted physicians to suggest potential beneficial effects of VD3A on skin pre-cancerous and cancerous lesions, with the focus primarily directed towards calcipotriol. Over the last decade, several clinical trials have emerged to investigate the efficacy of combining calcipotriol (0.005% foam or ointment) with 5-FU (1–5% cream) as a local chemotherapy for AK and SCC [281,282,283,284]. In general, they have established improved efficacy and accelerated onset of action through the induction of anti-tumor T-cell immunity in comparison to 5-FU monotherapy [285]. In addition, calcipotriol has been recognized as an enhancer of the PDT of AK [286]. Although vitamin D3 and VD3A have shown independent anti-cancer properties overall, the so-far established effects seem to be selective and related to VDR expression in the tumor cell line [287,288,289,290]. Therewith, Podgorska et al. have explained the lack of eloquent anti-tumor activity against melanoma in vitro. However, the same authors emphasized the sensitizing effect of calcitriol or calcidiol (Vitamin D3 active metabolite and precursor, respectively) on melanoma cancer cells to ionizing (proton beam) radiation [291]. While many researchers have attempted to nano-formulate VD3A in order to achieve improved, prolonged, and/or safer antipsoriatic efficacy [292,293,294], no such attempts were found in the direction of improved therapy of skin cancers.

6.7. Retinoids

Synthetic retinoids evolve throughout several generations, seeking enhanced chemical stability, selectivity to retinoid receptor(s) subtypes, improved tolerability, and higher skin bioavailability at the expense of limited percutaneous absorption for the topically active representatives [295,296]. Generations I, III, and IV are primarily designated for the treatment of acne, while Generation II (etretinate and acitretin) is established for the oral therapy of psoriasis [295,296,297,298]. All-trans retinoic acid—tretinoin (first generation), adapalene (third generation), and tazarotene (third generation)—has shown promising results in skin cancer treatment and prevention [296,298]. Tretinoin has been available on the market the longest, first as a solution and a cream for dermal use (Retin-A® 0.1%), and later as a gel, and as a microspheres-enriched modified-release gel (Retin-A micro® 0.04–0.1%) [299]; therefore, it has been investigated in the direction of skin cancer the most and is even used off-label for prevention [299]. The compound acts as a non-selective agonist of the retinoic acid receptors (RARs; isomers α, β, and γ) and the retinoid X receptors (RXRs; isomers α, β, and γ) [295,296]. Independently, tretinoin is proven to downregulate pro-inflammatory nuclear transcription factors [295]. Altogether, the topical use of tretinoin is found to result in the induction of keratinocyte apoptosis, the enhancement of cell turnover, the inhibition of carcinogenesis, and the suppression of tumor growth [296]. Data are available for the effectiveness of topical tretinoin in the treatment of AK, BD, SCC, and BCC [299,300,301]. The selective RAR-β/RAR-γ agonist adapalene (available as Differin® 0.1% cream or gel) is found to be efficient in reducing AK lesions and exerting anti-proliferative activity against melanoma cell lines [301,302,303,304]. Tazarotene (also a third-generation selective RAR-β/RAR-γ agonist; available as Tazorac®/Fabior® 0.1% cream, gel, or foam) has additionally shown activity against BCC [305,306]; however, a clinical trial investigating its activity in patients with BCC nevus syndrome has disputed potency for this indication [307]. The purely RAR-γ selective trifarotene (fourth generation; available as Aklief® 0.005% cream) is to date only suspected for its potential in skin cancers because of the mechanism of action [308]. Bexarotene, a third-generation RXR-selective agonist (referred to as a rexinoid representative), is available as Targetin® 1% gel and is indicated for cutaneous T-cell lymphoma (CTCL) [296,309]. Another rexinoid, alitretinoin (as a 0.1% gel), is approved by the FDA for the topical treatment of Kaposi’s sarcoma [310].
Retinoids as topical therapeutic agents suffer several disadvantages, such as high potency of causing skin irritability and dryness, chemical instability (photosensitivity of the earlier generations), or extreme lipophilicity (for example, log P of bexarotene is 6.9) [295,296,309]. Thus, they represent ideal candidates for prodrug and nano-formulation. However, since skin cancers are not their primary indication, very few of the innovative drug developments with retinoids address advancement in this clinical direction. Sallam et al. accomplished enhanced activity against SCC and CTCL with an increased skin deposition rate when conjugating bexarotene with hyaluronic acid [311]. Warda et al. obtained several bexarotene analogs with increased potency to inhibit CTCL cells by nitrogen substitution/hydroxylation/ring contraction [312]. Shah et al. achieved considerably improved photostability and skin tolerability of tretinoin (as compared to Retin-A® cream) when formulating it into SLNs and administering the drug-loaded particles in the form of a gel; however, the authors only suggest potential benefits in the topical therapy of skin cancers and pre-cancerous conditions [313].

6.8. Miscellaneous

Apart from the aforementioned APIs for the local chemotherapy of skin cancers and pre-cancerous lesions, which, one way or another, have reached clinical practice, there are numerous therapeutic entities in a research phase that are also showing promising results. These include repurposed antineoplastic agents, antibiotics, phytochemicals, inorganic nanoparticles, and others. The most recent developments rely on nanotechnology to maximize the efficiency and safety of the cutaneous application, leaning on all the arguments discussed in the Introductory Section of this paper. Also, an increasing number of researchers bet on dual or even triple combinations of nanotechnological approaches, an additional advanced drug delivery platform, and physical enhancers (e.g., iontophoresis, ultrasound, microneedles, etc.). Although the latter developments suggest much higher manufacturing and therapy costs, as well as a challenging reproducibility of the production technology, they hold the potential to solve more problems of the local chemotherapy of skin cancers [314]. Table 5 summarizes trends and achievements from the last few years.

7. Conclusions

Based on this review, local chemotherapy for skin pre-neoplastic and malignant lesions appears to be a rapidly evolving field. It is mainly driven by the high demand for non-invasive, more effective, targeted, safe, and patient-friendly therapeutic approaches. While conventional agents such as 5-FU, IMQ, tirbanibulin, IM, and diclofenac serve to date as clinical standards, their limitations (e.g., skin penetration, chemical stability, local tolerability, etc.) underscore the need for advanced delivery strategies. The various nanotechnological approaches offer enhanced skin permeation, improved drug stability, targeted delivery, and reduced systemic exposure. Numerous and diverse nano-scaled delivery platforms, including vesicular systems, SLNs, NLCs, polymeric, hybrid, and inorganic nano-carriers, have demonstrated promising preclinical and clinical advancements in the treatment of AK, SCC, BCC, and melanoma. Moreover, the repurposing of established APIs, along with the integration of natural compounds and prodrug strategies, expands the range of therapeutic options. For the sake of objectivity, challenges such as safety validation, large-scale manufacturing, and regulatory approval remain. As in many other therapeutic fields, the clinical translation of valuable laboratory findings requires a solid scientific background and further controlled clinical trials to support data for safety and efficacy.

Funding

This work was funded by Fund “Nauka” at the Medical University of Varna, Bulgaria, grant number Project No. 20016.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

During the preparation of this manuscript, the author used scispace.com for the purpose of data collection, as well as ChatGPT 4.0 for the purpose of withdrawing crucial points for the Conclusion section. The author have reviewed and edited the output and take full responsibility for the content of this publication.

Conflicts of Interest

The author declares no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
SCCSquamous cell carcinoma
NMSCsNon-melanoma skin cancers
BCCBasal cell carcinoma
UVUltraviolet
ROSReactive oxygen species
RNSReactive nitrogen species
HPVHuman papillomavirus
AKActinic keratosis
BDBowen’s disease
SCStratum corneum
EPREnhanced permeability and retention effect
PDTPhotodynamic therapy
5-FU5-Fluorouracil
APIActive pharmaceutical ingredient
DMSODimethylsulfoxide
SLNsSolid lipid nanoparticles
NLCsNanostructured lipid carriers
AgNPsSilver nanoparticles
AuNPsGold nanoparticles
STINGStimulator of Interferon Genes
BCSBiopharmaceutical classification system
IMQImiquimod
FDAUnited States Food and Drug Administration
EMAEuropean Medicines Agency
ADPAdenosine diphosphate ribose
COX-2Cyclooxygenase-2
PGE2Prostaglandin E2
IMIngenol mebutate
VD3AVitamin D3 analogs
VDRVitamin D receptor
RARsRetinoic acid receptors
RXRsRetinoid X receptors
CTCLCutaneous T-cell lymphoma
STAT3Signal Transducer and Activator of Transcription 3
siRNASmall interfering RNA

References

  1. Roky, A.H.; Islam, M.M.; Ahasan, A.M.F.; Mostaq, M.S.; Mahmud, M.Z.; Amin, M.N.; Mahmud, M.A. Overview of skin cancer types and prevalence rates across continents. Cancer Pathog. Ther. 2024, 3, 89–100. [Google Scholar] [CrossRef]
  2. Nakamura, A.; Kataoka, K.; Takatsuka, S.; Takenouchi, T. Aging trends in skin cancer: A long-term observational study in Japan. JAAD Int. 2023, 13, 32–34. [Google Scholar] [CrossRef]
  3. Saginala, K.; Barsouk, A.; Aluru, J.S.; Rawla, P.; Barsouk, A. Epidemiology of Melanoma. Med. Sci. 2021, 9, 63. [Google Scholar] [CrossRef] [PubMed]
  4. Davis, L.E.; Shalin, S.C.; Tackett, A.J. Current state of melanoma diagnosis and treatment. Cancer Biol. Ther. 2019, 20, 1366–1379. [Google Scholar] [CrossRef] [PubMed]
  5. Zeng, L.; Gowda, B.H.J.; Ahmed, M.G.; Abourehab, M.A.S.; Chen, Z.-S.; Zhang, C.; Li, J.; Kesharwani, P. Advancements in nanoparticle-based treatment approaches for skin cancer therapy. Mol. Cancer 2023, 22, 10. [Google Scholar] [CrossRef]
  6. Attal, Z.G.; Shalata, W.; Soklakova, A.; Tourkey, L.; Shalata, S.; Abu Saleh, O.; Abu Salamah, F.; Alatawneh, I.; Yakobson, A. Advanced and Metastatic Non-Melanoma Skin Cancer: Epidemiology, Risk Factors, Clinical Features, and Treatment Options. Biomedicines 2024, 12, 1448. [Google Scholar] [CrossRef]
  7. Newlands, C.; Currie, R.; Memon, A.; Whitaker, S.; Woolford, T. Non-melanoma skin cancer: United Kingdom National Multidisciplinary Guidelines. J. Laryngol. Otol. 2016, 130, S125–S132. [Google Scholar] [CrossRef] [PubMed]
  8. Khayyati Kohnehshahri, M.; Sarkesh, A.; Mohamed Khosroshahi, L.; HajiEsmailPoor, Z.; Aghebati-Maleki, A.; Yousefi, M.; Aghebati-Maleki, L. Current status of skin cancers with a focus on immunology and immunotherapy. Cancer Cell Int. 2023, 23, 174. [Google Scholar] [CrossRef]
  9. Tang, X.; Yang, T.; Yu, D.; Xiong, H.; Zhang, S. Current insights and future perspectives of ultraviolet radiation (UV) exposure: Friends and foes to the skin and beyond the skin. Environ. Int. 2024, 185, 108535. [Google Scholar] [CrossRef]
  10. Norval, M. The mechanisms and consequences of ultraviolet-induced immunosuppression. Prog. Biophys. Mol. Biol. 2006, 92, 108–118. [Google Scholar] [CrossRef]
  11. Liu, H.M.; Cheng, M.Y.; Xun, M.H.; Zhao, Z.W.; Zhang, Y.; Tang, W.; Cheng, J.; Ni, J.; Wang, W. Possible Mechanisms of Oxidative Stress-Induced Skin Cellular Senescence, Inflammation, and Cancer and the Therapeutic Potential of Plant Polyphenols. Int. J. Mol. Sci. 2023, 24, 3755. [Google Scholar] [CrossRef] [PubMed]
  12. Rahman, I. Oxidative Stress in the Pathogenesis of Chronic Obstructive Pulmonary Disease. In Cigarette Smoke and Oxidative Stress; Springer: Berlin/Heidelberg, Germany, 2006; pp. 165–197. [Google Scholar] [CrossRef]
  13. Adnan, M.; Akhter, M.H.; Afzal, O.; Altamimi, A.S.A.; Ahmad, I.; Alossaimi, M.A.; Jaremko, M.; Emwas, A.-H.; Haider, T.; Haider, M.F. Exploring Nanocarriers as Treatment Modalities for Skin Cancer. Molecules 2023, 28, 5905. [Google Scholar] [CrossRef]
  14. Prohic, A. Precancerous Skin Lesions. In Dermatovenerology Textbook; Springer: Berlin/Heidelberg, Germany, 2024; pp. 497–506. [Google Scholar] [CrossRef]
  15. Hidalgo, L.; Saldías-Fuentes, C.; Carrasco, K.; Halpern, A.C.; Mao, J.J.; Navarrete-Dechent, C. Complementary and alternative therapies in skin cancer: A literature review of biologically active compounds. Dermatol. Ther. 2022, 35, e15842. [Google Scholar] [CrossRef]
  16. Jadhav, L.A.; Mandlik, S.K. Nanocarriers in skin cancer treatment: Emerging drug delivery approaches and innovations. Nano TransMed 2025, 4, 100068. [Google Scholar] [CrossRef]
  17. Chang, J.; Yu, B.; Saltzman, W.M.; Girardi, M. Nanoparticles as a Therapeutic Delivery System for Skin Cancer Prevention and Treatment. JID Innov. 2023, 3, 100197. [Google Scholar] [CrossRef]
  18. Maru, G.B.; Gandhi, K.; Ramchandani, A.; Kumar, G. The role of inflammation in skin cancer. Adv. Exp. Med. Biol. 2014, 816, 437–469. [Google Scholar] [CrossRef]
  19. Rangwala, S.; Tsai, K.Y. Roles of the immune system in skin cancer. Br. J. Dermatol. 2011, 165, 953–965. [Google Scholar] [CrossRef]
  20. Huang, Y.; Carlsson, L.; Jogeland, K.; Samuelsson, M.; Larsson, L.; Jonsborg, C. Management of complications after skin surgery relevant for melanoma in the trunk and extremities during the COVID-19 pandemic: A case series report. World J. Surg. Oncol. 2023, 21, 280. [Google Scholar] [CrossRef]
  21. Khan, S.U.; Ullah, M.; Saeed, S.; Saleh, E.A.M.; Kassem, A.F.; Arbi, F.M.; Wahab, A.; Rehman, M.; ur Rehman, K.; Khan, D.; et al. Nanotherapeutic approaches for transdermal drug delivery systems and their biomedical applications. Eur. Polym. J. 2024, 207, 112819. [Google Scholar] [CrossRef]
  22. Leong, M.Y.; Kong, Y.L.; Burgess, K.; Wong, W.F.; Sethi, G.; Looi, C.Y. Recent development of nanomaterials for transdermal drug delivery. Biomedicines 2023, 11, 1124. [Google Scholar] [CrossRef] [PubMed]
  23. Mearaj, S.; Kim, T.M.; Choi, J.W. Photoprotective and bioinspired cerium-encapsulating lignin nanocapsules for multifunctional UV protection applications. Chem. Eng. J. 2024, 501, 157668. [Google Scholar] [CrossRef]
  24. Smijs, T.G.; Pavel, S. Titanium dioxide and zinc oxide nanoparticles in sunscreens: Focus on their safety and effectiveness. Nanotechnol. Sci. Appl. 2011, 4, 95–112. [Google Scholar] [CrossRef]
  25. Yabe, S.; Sato, T. Cerium oxide for sunscreen cosmetics. J. Solid. State Chem. 2003, 171, 7–11. [Google Scholar] [CrossRef]
  26. Brito, S.; Baek, M.; Bin, B.-H. Skin Structure, Physiology, and Pathology in Topical and Transdermal Drug Delivery. Pharmaceutics 2024, 16, 1403. [Google Scholar] [CrossRef]
  27. Gallo, R.L. Human Skin Is the Largest Epithelial Surface for Interaction with Microbes. J. Investig. Dermatol. 2017, 137, 1213–1214. [Google Scholar] [CrossRef] [PubMed]
  28. McBride, A.; Bargmann, S.; Pond, D.; Limbert, G. Thermoelastic Modelling of the Skin at Finite Deformations. J. Therm. Biol. 2016, 62, 201–209. [Google Scholar] [CrossRef] [PubMed]
  29. Dermitzakis, I.; Chatzi, D.; Kyriakoudi, S.A.; Evangelidis, N.; Vakirlis, E.; Meditskou, S.; Theotokis, P.; Manthou, M.E. Skin Development and Disease: A Molecular Perspective. Curr. Issues Mol. Biol. 2024, 46, 8239–8267. [Google Scholar] [CrossRef] [PubMed]
  30. Lukić, M.; Pantelić, I.; Savić, S.D. Towards Optimal pH of the Skin and Topical Formulations: From the Current State of the Art to Tailored Products. Cosmetics 2021, 8, 69. [Google Scholar] [CrossRef]
  31. Kuo, S.-H.; Shen, C.-J.; Shen, C.-F.; Cheng, C.-M. Role of pH Value in Clinically Relevant Diagnosis. Diagnostics 2020, 10, 107. [Google Scholar] [CrossRef]
  32. Sanchez, M.M.; Tonmoy, T.I.; Park, B.H.; Morgan, J.T. Development of a Vascularized Human Skin Equivalent with Hypodermis for Photoaging Studies. Biomolecules 2022, 12, 1828. [Google Scholar] [CrossRef]
  33. Elias, P.M. Structure and Function of the Stratum Corneum Extracellular Matrix. J. Investig. Dermatol. 2012, 132, 2131–2133. [Google Scholar] [CrossRef]
  34. Santiago, J.L.; Muñoz-Rodriguez, J.R.; Cruz-Morcillo, M.A.d.l.; Villar-Rodriguez, C.; Gonzalez-Lopez, L.; Aguado, C.; Nuncia-Cantarero, M.; Redondo-Calvo, F.J.; Perez-Ortiz, J.M.; Galan-Moya, E.M. Characterization of Permeability Barrier Dysfunction in a Murine Model of Cutaneous Field Cancerization Following Chronic UV-B Irradiation: Implications for the Pathogenesis of Skin Cancer. Cancers 2021, 13, 3935. [Google Scholar] [CrossRef]
  35. Ishitsuka, Y.; Roop, D.R. Loricrin: Past, Present, and Future. Int. J. Mol. Sci. 2020, 21, 2271. [Google Scholar] [CrossRef] [PubMed]
  36. Fujii, M. The Pathogenic and Therapeutic Implications of Ceramide Abnormalities in Atopic Dermatitis. Cells 2021, 10, 2386. [Google Scholar] [CrossRef] [PubMed]
  37. Alkilani, A.Z.; McCrudden, M.T.; Donnelly, R.F. Transdermal Drug Delivery: Innovative Pharmaceutical Developments Based on Disruption of the Barrier Properties of the Stratum Corneum. Pharmaceutics 2015, 7, 438–470. [Google Scholar] [CrossRef] [PubMed]
  38. Kougkolos, G.; Laudebat, L.; Dinculescu, S.; Simon, J.; Golzio, M.; Valdez-Nava, Z.; Flahaut, E. Skin Electroporation for Transdermal Drug Delivery: Electrical Measurements, Numerical Model and Molecule Delivery. J. Control Release 2024, 367, 235–247. [Google Scholar] [CrossRef]
  39. Ogórek, K.; Nowak, K.; Wadych, E.; Ruzik, L.; Timerbaev, A.R.; Matczuk, M. Are We Ready to Measure Skin Permeation of Modern Antiaging GHK–Cu Tripeptide Encapsulated in Liposomes? Molecules 2025, 30, 136. [Google Scholar] [CrossRef]
  40. Ramadon, D.; McCrudden, M.T.C.; Courtenay, A.J.; Donnelly, R.F. Enhancement Strategies for Transdermal Drug Delivery Systems: Current Trends and Applications. Drug Deliv. Transl. Res. 2022, 12, 758–791. [Google Scholar] [CrossRef]
  41. Jain, S.K.; Verma, A.; Jain, A.; Hurkat, P. Transfollicular Drug Delivery: Current Perspectives. Res. Rep. Transdermal Drug Deliv. 2016, 5, 1–17. [Google Scholar] [CrossRef]
  42. Meidan, V.M.; Bonner, M.C.; Michniak, B.B. Transfollicular Drug Delivery—Is it a Reality? Int. J. Pharm. 2005, 306, 1–14. [Google Scholar] [CrossRef]
  43. Rancan, F.; Vogt, A. Getting Under the Skin: What Is the Potential of the Transfollicular Route in Drug Delivery? Ther. Deliv. 2014, 5, 875–877. [Google Scholar] [CrossRef]
  44. Supe, S.; Takudage, P. Methods for Evaluating Penetration of Drug into the Skin: A Review. Ski. Res. Technol. 2020, 27, 299–308. [Google Scholar] [CrossRef]
  45. Bolzinger, M.-A.; Briançon, S.; Pelletier, J.; Chevalier, Y. Penetration of Drugs through Skin, A Complex Rate-controlling Membrane. Curr. Opin. Colloid. Interface Sci. 2012, 17, 156–165. [Google Scholar] [CrossRef]
  46. Farage, M.A.; Miller, K.W.; Elsner, P.; Maibach, H.I. Characteristics of the Aging Skin. Adv. Wound Care 2013, 2, 5–10. [Google Scholar] [CrossRef] [PubMed]
  47. Russell-Goldman, E.; Murphy, G.F. The Pathobiology of Skin Aging: New Insights into an Old Dilemma. Am. J. Pathol. 2020, 190, 1356–1369. [Google Scholar] [CrossRef] [PubMed]
  48. Wang, Z.; Man, M.Q.; Li, T.; Elias, P.M.; Mauro, T.M. Aging-associated alterations in epidermal function and their clinical significance. Aging 2020, 12, 5551–5565. [Google Scholar] [CrossRef]
  49. White-Chu, E.F.; Reddy, M. Dry skin in the elderly: Complexities of a common problem. Clin. Dermatol. 2011, 29, 37–42. [Google Scholar] [CrossRef]
  50. Ribeiro, M.; Simões, M.; Vitorino, C.; Mascarenhas-Melo, F. Hydrogels in Cutaneous Wound Healing: Insights into Characterization, Properties, Formulation and Therapeutic Potential. Gels 2024, 10, 188. [Google Scholar] [CrossRef]
  51. Souto, E.B.; Fangueiro, J.F.; Fernandes, A.R.; Cano, A.; Sanchez-Lopez, E.; Garcia, M.L.; Severino, P.; Paganelli, M.O.; Chaud, M.V.; Silva, A.M. Physicochemical and Biopharmaceutical Aspects Influencing Skin Permeation and Role of SLN and NLC for Skin Drug Delivery. Heliyon 2022, 8, e08938. [Google Scholar] [CrossRef]
  52. Rawlings, A.V.; Matts, P.J. Stratum Corneum Moisturization at the Molecular Level: An Update in Relation to the Dry Skin Cycle. J. Investig. Dermatol. 2005, 124, 1099–1110. [Google Scholar] [CrossRef]
  53. Załęcki, P.; Rogowska, K.; Wąs, P.; Łuczak, K.; Wysocka, M.; Nowicka, D. Impact of Lifestyle on Differences in Skin Hydra-Tion of Selected Body Areas in Young Women. Cosmetics 2024, 11, 13. [Google Scholar] [CrossRef]
  54. Gidado, I.; Qassem, M.; Triantis, I.; Kyriacou, P. Review of Advances in the Measurement of Skin Hydration Based on Sensing of Optical and Electrical Tissue Properties. Sensors 2022, 22, 7151. [Google Scholar] [CrossRef]
  55. Goad, N.; Gawkrodger, D. Ambient Humidity and the Skin: The Impact of Air Humidity in Healthy and Diseased States. J. Eur. Acad. Dermatol. Venereol. 2016, 30, 1285–1294. [Google Scholar] [CrossRef] [PubMed]
  56. Walters, K.; Lane, M. Dermal and Transdermal Drug Delivery Systems. In Dermal Drug Delivery; CRC Press: Boca Raton, FL, USA, 2020; pp. 1–60. [Google Scholar]
  57. David, P.; Singh, S.; Ankar, R.A. Comprehensive Overview of Skin Complications in Diabetes and Their Prevention. Cureus 2023, 15, e38961. [Google Scholar] [CrossRef]
  58. Giakoumaki, M.; Lambrou, G.I.; Vlachodimitropoulos, D.; Tagka, A.; Vitsos, A.; Kyriazi, M.; Dimakopoulou, A.; Anagnostou, V.; Karasmani, M.; Deli, H. Type I Diabetes Mellitus Suppresses Experimental Skin Carcinogenesis. Cancers 2024, 16, 1507. [Google Scholar] [CrossRef]
  59. Safer, J. Thyroid Hormone Action on Skin. Dermatoendocrinology 2011, 3, 211–215. [Google Scholar] [CrossRef]
  60. Gratton, R.; Del Vecchio, C.; Zupin, L.; Crovella, S. Unraveling the Role of Sex Hormones on Keratinocyte Functions in Human Inflammatory Skin Diseases. Int. J. Mol. Sci. 2022, 23, 3132. [Google Scholar] [CrossRef] [PubMed]
  61. Kim, H.J.; Kim, Y.H. Exploring Acne Treatments: From Pathophysiological Mechanisms to Emerging Therapies. Int. J. Mol. Sci. 2024, 25, 5302. [Google Scholar] [CrossRef]
  62. Gruber, J.V.; Stojkoska, V.; Riemer, J. Retinol Has a Skin Dehydrating Effect That Can Be Improved by a Mixture of Wa-Ter-Soluble Polysaccharides. Cosmetics 2020, 7, 80. [Google Scholar] [CrossRef]
  63. Kowalska, J.; Wrześniok, D. Skin-Related Adverse Reactions Induced by Oral Antidiabetic Drugs—A Review of Literature and Case Reports. Pharmaceuticals 2024, 17, 847. [Google Scholar] [CrossRef] [PubMed]
  64. Pyo, S.; Maibach, H. Skin Metabolism: Relevance of Skin Enzymes for Rational Drug Design. Ski. Pharmacol. Physiol. 2019, 32, 283–294. [Google Scholar] [CrossRef]
  65. Suresh, P.K. Enhanced Permeation Retention Effect-Modeling and Imaging Approaches for Nanoparticle-Mediated Anti-Cancer Diagnostics or Therapy. Lett. Drug Des. Discov. 2024, 21, 3633–3638. [Google Scholar] [CrossRef]
  66. Serzants, R.; Svalbe, B.; Cesnokova, I.; Stelfa, G.; Sizovs, A. Exploring the Limits of EPR-Driven Tumor Accumulation with Non-Opsonizing Nanomaterials. bioRxiv 2024. [Google Scholar] [CrossRef]
  67. Vijaykumar, V.; Saikiran, M.; Bharathy, V.; Ubaidulla, U. Formulation Challenges in Dermal Drug Delivery Systems: A Comprehensive Review of Physicochemical Properties and Advanced Delivery Strategies. Int. J. Drug Deliv. Technol. 2024, 14, 1124–1129. [Google Scholar] [CrossRef]
  68. Czyrski, G.S.; Hjort, M.K.F.; Rades, T.; Heinz, A. Comparing Effects of Terpene-Based Deep Eutectic Solvent and Solid Microneedles on Skin Permeation of Drugs with Varying Lipophilicity. Eur. J. Pharm. Biopharm. 2024, 205, 114576. [Google Scholar] [CrossRef]
  69. Kilian, D.; Lemmer, H.J.R.; Gerber, M.; Du Preez, J.L.; Plessis, J. Exploratory Data Analysis of the Dependencies between Skin Permeability, Molecular Weight and Log P. Die Pharm. 2016, 71, 311–319. [Google Scholar] [CrossRef]
  70. Goold, S.R.; Raddi, R.M.; Voelz, V.A. Expanded ensemble predictions of toluene–water partition coefficients in the SAMPL9 log P challenge. Phys. Chem. Chem. Phys. 2025, 27, 6005–6013. [Google Scholar] [CrossRef] [PubMed]
  71. Algotsson, J.; Jönsson, P.; Forsman, J.; Topgaard, D.; Söderman, O. Intermolecular Interactions Play a Role in the Distribution and Transport of Charged Contrast Agents in a Cartilage Model. PLoS ONE 2019, 14, e0215047. [Google Scholar] [CrossRef]
  72. Balouch, M.; Storchmannová, K.; Štěpánek, F.; Berka, K. Computational Prodrug Design Methodology for Liposome Formulability Enhancement of Small-Molecule APIs. Mol. Pharm. 2023, 20, 2119–2127. [Google Scholar] [CrossRef] [PubMed]
  73. Gavhane, S.A.; Somwanshi, S.B. Prodrug: Approach to Better Drug Delivery; IIP Series; IIP: Chikkamagaluru, India, 2024; pp. 56–61. [Google Scholar]
  74. Sahu, S.K.; Singh, A.; Verma, P.; Chaurasiya, S. Prodrug: A Brief Concept; IIP Series; IIP: Chikkamagaluru, India, 2024; pp. 247–256. [Google Scholar]
  75. Zeng, Y.; Ma, M.; Chen, Y.; Xie, H.; Ding, P.; Zhang, K. Enhancing Skin Delivery of Tranexamic Acid via Esterification: Synthesis and Evaluation of Alkyl Ester Derivatives. RSC Adv. 2024, 14, 34996–35004. [Google Scholar] [CrossRef] [PubMed]
  76. Wu, F.; Ni, F.; Yao, J.; Huang, C.; Zhao, Y. Synthesis of Phosphoramidate Prodrugs of Phenolic Natural Products and Drugs by Ester Exchange. Synthesis 2022, 54, 3005–3014. [Google Scholar] [CrossRef]
  77. Suppasansatorn, P.; Wang, G.; Conway, B.R.; Wang, W.; Wang, Y. Skin Delivery Potency and Antitumor Activities of Temozolomide Ester Prodrugs. Cancer Lett. 2006, 244, 42–52. [Google Scholar] [CrossRef]
  78. Filonenko, E.; Kaprin, A.; Urlova, A.; Grigorievykh, N.; Ivanova-Radkevich, V. Topical 5-Aminolevulinic Acid-Mediated Photodynamic Therapy for Basal Cell Carcinoma. Photodiagnosis Photodyn. Ther. 2020, 30, 101644. [Google Scholar] [CrossRef] [PubMed]
  79. Gold, M. Therapeutic and Aesthetic Uses of Photodynamic Therapy Part Five of a Five-Part Series: ALA-PDT in Clinical Practice How One Clinician Performs This Procedure. J. Clin. Aesthet. Dermatol. 2009, 2, 32–35. [Google Scholar] [PubMed]
  80. Champeau, M.; Vignoud, S.; Mortier, L.; Mordon, S. Photodynamic Therapy for Skin Cancer: How to Enhance Drug Penetration? J. Photochem. Photobiol. B 2019, 197, 111544. [Google Scholar] [CrossRef] [PubMed]
  81. Winkler, A.; Müller-Goymann, C. Comparative Permeation Studies for Delta-Aminolevulinic Acid and Its n-Butylester through Stratum Corneum and Artificial Skin Constructs. Eur. J. Pharm. Biopharm. 2002, 53, 281–287. [Google Scholar] [CrossRef]
  82. Neittaanmäki-Perttu, N.; Grönroos, M.; Karppinen, T.T.; Tani, T.T.; Snellman, E. Hexyl-5-aminolaevulinate 0·2% vs. Methyl-5-aminolaevulinate 16% Daylight Photodynamic Therapy for Treatment of Actinic Keratoses: Results of a Randomized Double-blinded Pilot Trial. Br. J. Dermatol. 2016, 174, 427–429. [Google Scholar] [CrossRef]
  83. Fang, J.; Leu, Y.L. Prodrug Strategy for Enhancing Drug Delivery via Skin. Curr. Drug Discov. Technol. 2006, 3, 211–224. [Google Scholar] [CrossRef]
  84. Quigley, J.; Lloyd, D. A Topological Study of Prodrugs of 5-Fluorouracil. Int. J. Pharm. 2002, 231, 241–251. [Google Scholar] [CrossRef]
  85. Patrick, A.I.; Beall, H.D.; Gilroy, P.; Sloan, K.B. Effect of Vehicles on Topical Delivery of 5-Fluorouracil (5FU) by 1-Acyl-5FU Prodrugs. Int. J. Pharm. 1997, 154, 39–48. [Google Scholar] [CrossRef]
  86. Jacobson, E.; Kim, H.; Kim, M.; Wondrak, G.; Jacobson, M. Developing Topical Prodrugs for Skin Cancer Prevention. In Fundamentals of Cancer Prevention; Alberts, D., Hess, L., Eds.; Springer: Berlin/Heidelberg, Germany, 2005; pp. 139–160. [Google Scholar]
  87. Sharma, A.; Sharma, D.; Baldi, A.; Jyoti, K.; Chandra, R.; Madan, J. Imiquimod-Oleic Acid Prodrug-Loaded Cream Reduced Drug Crystallinity and Induced Indistinguishable Cytotoxicity and Apoptosis in Mice Melanoma Tumour. J. Microencapsul. 2019, 36, 759–774. [Google Scholar] [CrossRef]
  88. Vaienti, S.; Calzari, P.; Nazzaro, G. Topical Treatment of Melanoma In Situ, Lentigo Maligna, and Lentigo Maligna Melanoma with Imiquimod Cream: A Systematic Review of the Literature. Dermatol. Ther. 2023, 13, 2187–2215. [Google Scholar] [CrossRef]
  89. Lein, A.; Oussoren, C. Dermal. In Practical Pharmaceutics; Springer International Publishing Switzerland: Cham, Switzerland, 2015; pp. 229–263. [Google Scholar]
  90. Ahuja, K.; An, M.; Lio, P. A Brief Review of Vehicles for Topical Therapies. Ski. Pharmacol. Physiol. 2025, 37, 104–108. [Google Scholar] [CrossRef]
  91. Herbig, M.E.; Evers, D.-H.; Gorissen, S.; Köllmer, M. Rational Design of Topical Semi-Solid Dosage Forms-How Far Are We? Pharmaceutics 2023, 15, 1822. [Google Scholar] [CrossRef]
  92. Kim, S.; Day, C.; Song, Y.; Holmes, A.; Garg, S. Innovative Topical Patches for Non-Melanoma Skin Cancer: Current Challenges and Key Formulation Considerations. Pharmaceutics 2023, 15, 2577. [Google Scholar] [CrossRef]
  93. Lei, Q.; He, D.; Ding, L.; Kong, F.; He, P.; Huang, J.; Guo, J.; Brinker, C.J.; Luo, G.; Zhu, W.; et al. Microneedle Patches Integrated with Biomineralized Melanin Nanoparticles for Simultaneous Skin Tumor Photothermal Therapy and Wound Healing. Adv. Funct. Mater. 2022, 32, 2113269. [Google Scholar] [CrossRef]
  94. Dragicevic, N.; Predic Atkinson, J.; Maibach, H.I. Chemical Penetration Enhancers: Classification and Mode of Action; Springer: Berlin/Heidelberg, Germany, 2015. [Google Scholar]
  95. Špaglová, M.; Žigrayová, D.; Krchňák, D. Development Strategy of Dermal and Transdermal Formulation: Synergistic Effect of Chemical Penetration Enhancers. Acta Med. Bulg. 2024, 51, 171–181. [Google Scholar] [CrossRef]
  96. Borude, P.D. A Review Article on Permeability Enhancement. Int. J. Res. Appl. Sci. Eng. Technol. 2024, 12, 2301–2305. [Google Scholar] [CrossRef]
  97. Greve, T.M.; Andersen, K.B.; Nielsen, O.F. Penetration Mechanism of Dimethyl Sulfoxide in Human and Pig Ear Skin: An ATR-FTIR and near-FT Raman Spectroscopic in Vivo and in Vitro Study. Spectroscopy 2008, 22, 405–417. [Google Scholar] [CrossRef]
  98. Kulkarni, V.S.; Shaw, C. Use of Polymers and Thickeners in Semisolid and Liquid Formulations. In Essential Chemistry for Formulators of Semisolid and Liquid Dosages; Kulkarni, V.S., Shaw, C., Eds.; Academic Press: Cambridge, MA, USA, 2016; pp. 43–69. [Google Scholar]
  99. Jampílek, J. Azone® and Its Analogues as Penetration Enhancers. In Percutaneous Penetration Enhancers Chemical Methods in Penetration Enhancement; Springer: London, NY, USA, 2015; pp. 69–105. [Google Scholar]
  100. Singh, S.; Mazumder, R.; PADHI, S.; Mishra, R.; Kumar, V. Unlocking Skin Barriers: Applications and Properties of Natural Permeation Enhancers. J. Nat. Remedies 2024, 24, 2599–2624. [Google Scholar] [CrossRef]
  101. Hmingthansanga, V.; Singh, N.; Banerjee, S.; Manickam, S.; Velayutham, R.; Natesan, S. Improved Topical Drug Delivery: Role of Permeation Enhancers and Advanced Approaches. Pharmaceutics 2022, 14, 2818. [Google Scholar] [CrossRef] [PubMed]
  102. Dudhat, K. Emerging Trends in Transdermal Drug Delivery: Nanoparticle Formulations and Technologies for Enhanced Skin Penetration and Drug Efficiency. Pharm. Nanotechnol. 2024, 13, 1–21. [Google Scholar] [CrossRef]
  103. Lalotra, A.S.; Singh, V.; Khurana, B.; Agrawal, S.; Shrestha, S.; Arora, D. A Comprehensive Review on Nanotechnology-Based Innovations in Topical Drug Delivery for the Treatment of Skin Cancer. Curr. Pharm. Des. 2020, 26, 5720–5731. [Google Scholar] [CrossRef]
  104. Krishnan, V.; Mitragotri, S. Nanoparticles for Topical Drug Delivery: Potential for Skin Cancer Treatment. Adv. Drug Deliv. Rev. 2020, 153, 87–108. [Google Scholar] [CrossRef] [PubMed]
  105. Nsairat, H.; Khater, D.; Sayed, U.; Odeh, F.; Al Bawab, A.; Alshaer, W. Liposomes: Structure, Composition, Types, and Clinical Applications. Heliyon 2022, 8, e09394. [Google Scholar] [CrossRef]
  106. Sercombe, L.; Veerati, T.; Moheimani, F.; Wu, S.; Sood, A.; Hua, S. Advances and Challenges of Liposome Assisted Drug Delivery. Front. Pharmacol. 2015, 6, 286. [Google Scholar] [CrossRef] [PubMed]
  107. Guillot, A.J.; Martínez-Navarrete, M.; Garrigues, T.M.; Melero, A. Skin Drug Delivery Using Lipid Vesicles: A Starting Guideline for Their Development. J. Control. Release 2023, 355, 624–654. [Google Scholar] [CrossRef]
  108. Gugleva, V.; Ivanova, N.; Sotirova, Y.; Andonova, V. Dermal Drug Delivery of Phytochemicals with Phenolic Structure via Lipid-Based Nanotechnologies. Pharmaceuticals 2021, 14, 837. [Google Scholar] [CrossRef]
  109. Zhan, B.; Wang, J.; Li, H.; Xiao, K.; Fang, X.; Shi, Y.; Jia, Y. Ethosomes: A Promising Drug Delivery Platform for Transdermal Application. Chemistry 2024, 6, 993–1019. [Google Scholar] [CrossRef]
  110. Musielak, E.; Krajka-Kuźniak, V. Liposomes and Ethosomes: Comparative Potential in Enhancing Skin Permeability for Therapeutic and Cosmetic Applications. Cosmetics 2024, 11, 191. [Google Scholar] [CrossRef]
  111. Matharoo, N.; Mohd, H.; Michniak-Kohn, B. Transferosomes as a Transdermal Drug Delivery System: Dermal Kinetics and Recent Developments. WIREs Nanomed. Nanobiotechnol. 2024, 16, e1918. [Google Scholar] [CrossRef]
  112. Fernández-García, R.; Lalatsa, A.; Statts, L.; Bolás-Fernández, F.; Ballesteros, M.P.; Serrano, D.R. Transferosomes as Nanocarriers for Drugs across the Skin: Quality by Design from Lab to Industrial Scale. Int. J. Pharm. 2020, 573, 118817. [Google Scholar] [CrossRef]
  113. Muzzalupo, R.; Tavano, L. Niosomal Drug Delivery for Transdermal Targeting: Recent Advances. Res. Rep. Transdermal Drug Deliv. 2015, 4, 23–33. [Google Scholar] [CrossRef]
  114. Kaur, P.; Kriplani, P. Quality by Design for Niosome-Based Nanocarriers to Improve Transdermal Drug Delivery from Lab to Industry. Int. J. Pharm. 2024, 666, 124747. [Google Scholar] [CrossRef] [PubMed]
  115. Oliveira, C.A.; Forster, C.; Feitosa, V.; Baby, A.R.; Léo, P.; Rangel-Yagui, C.O. Catalase-Loaded Polymersomes as a Promising Safe Ingredient to Active Photoprotection. J. Photochem. Photobiol. 2021, 7, 100056. [Google Scholar] [CrossRef]
  116. Fonseca, M.; Jarak, I.; Victor, F.; Domingues, C.; Veiga, F.; Figueiras, A. Polymersomes as the Next Attractive Generation of Drug Delivery Systems: Definition, Synthesis and Applications. Materials 2024, 17, 319. [Google Scholar] [CrossRef]
  117. Kotha, R.; Kara, D.; Roychowdhury, R.; Tanvi, K.; Rathnanand, M. Polymersomes Based Versatile Nanoplatforms for Controlled Drug Delivery and Imaging. Adv. Pharm. Bull. 2023, 13, 218–232. [Google Scholar] [CrossRef]
  118. Hua, C.; Qiu, L. Polymersomes for Therapeutic Protein and Peptide Delivery: Towards Better Loading Properties. Int. J. Nanomed. 2024, 19, 2317–2340. [Google Scholar] [CrossRef]
  119. Jain, S.; Tripathi, S.; Tripathi, P.K. Invasomes: Potential Vesicular Systems for Transdermal Delivery of Drug Molecules. J. Drug Deliv. Sci. Technol. 2021, 61, 102166. [Google Scholar] [CrossRef]
  120. Preeti, P.; Puri, D.; Singh, S. Invasomes: An Artificial Vesicle Nanocarrier to Enhance Transdermal Drug Delivery. Curr. Nanomed. 2024, 15, 1–15. [Google Scholar] [CrossRef]
  121. Afreen, U.; Shailaja, A.K. Pharmacosomes and Emulsomes: An Emerging Novel Vesicular Drug Delivery System. Glob. J. Anesthesia Pain Med. 2020, 3, 287–297. [Google Scholar] [CrossRef]
  122. Rana, P.; Mahajan, A.; Singh, D.; Singh, K. Pharmacosomes: A Versatile Delivery System for Problematic Molecules. Curr. Nanomed. 2021, 11, 82–90. [Google Scholar] [CrossRef]
  123. Gaikwad, S.S.; Morade, Y.Y.; Kothule, A.M.; Kshirsagar, S.J.; Laddha, U.D.; Salunkhe, K.S. Overview of Phytosomes in Treating Cancer: Advancement, Challenges, and Future Outlook. Heliyon 2023, 9, e16561. [Google Scholar] [CrossRef]
  124. Barani, M.; Sangiovanni, E.; Angarano, M.; Rajizadeh, M.; Mehrabani, M.; Piazza, S.; Gangadharappa, H.; Pardakhty, A.; Mehrbani, M.; Dell’Agli, M.; et al. Phytosomes as Innovative Delivery Systems for Phytochemicals: A Comprehensive Review of Literature. Int. J. Nanomed. 2021, 16, 6983–7022. [Google Scholar] [CrossRef]
  125. Nath, A.G.; Dubey, P.; Kumar, A.; Vaiphei, K.K.; Rosenholm, J.M.; Bansal, K.K.; Gulbake, A. Recent Advances in the Use of Cubosomes as Drug Carriers with Special Emphasis on Topical Applications. J. Lipids 2024, 2024, 2683466. [Google Scholar] [CrossRef]
  126. Marson, D.; Aulic, S.; Laurini, E.; Pricl, S. Chapter 9—Cubosomes: A Promising Vesicular System for Drug Delivery. In Systems of Nanovesicular Drug Delivery; Nayak, A.K., Hasnain, M.S., Aminabhavi, T.M., Torchilin, V.P., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 129–145. [Google Scholar]
  127. Jha, A.; Kumar, M.; Bharti, K.; Mishra, B. Chapter 17—Glycerosomes: A New Tool for Effective Drug Delivery. In Systems of Nanovesicular Drug Delivery; Nayak, A.K., Hasnain, M.S., Aminabhavi, T.M., Torchilin, V.P., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 277–291. [Google Scholar]
  128. Manca, M.L.; Zaru, M.; Manconi, M.; Lai, F.; Valenti, D.; Sinico, C.; Fadda, A.M. Glycerosomes: A New Tool for Effective Dermal and Transdermal Drug Delivery. Int. J. Pharm. 2013, 455, 66–74. [Google Scholar] [CrossRef] [PubMed]
  129. Mohammed, L.; Nourddine, H.; Saad, E.F.; Abdelali, D.; Hamid, R. Chitosan-Covered Liposomes as a Promising Drug Transporter: Nanoscale Investigations. RSC Adv. 2021, 11, 1503–1516. [Google Scholar] [CrossRef] [PubMed]
  130. Gaynanova, G.; Vasileva, L.; Kashapov, R.; Kuznetsova, D.; Kushnazarova, R.; Tyryshkina, A.; Vasilieva, E.; Petrov, K.; Zakharova, L.; Sinyashin, O. Self-Assembling Drug Formulations with Tunable Permeability and Biodegradability. Molecules 2021, 26, 6786. [Google Scholar] [CrossRef] [PubMed]
  131. Sebaaly, C.; Trifan, A.; Sieniawska, E.; Greige-Gerges, H. Chitosan-Coating Effect on the Characteristics of Liposomes: A Focus on Bioactive Compounds and Essential Oils: A Review. Processes 2021, 9, 445. [Google Scholar] [CrossRef]
  132. Colino, C.I.; Velez Gomez, D.; Alonso Horcajo, E.; Gutierrez-Millan, C. A Comparative Study of Liposomes and Chitosomes for Topical Quercetin Antioxidant Therapy. J. Drug Deliv. Sci. Technol. 2022, 68, 103094. [Google Scholar] [CrossRef]
  133. Esposto, B.S.; Jauregi, P.; Tapia-Blácido, D.R.; Martelli-Tosi, M. Liposomes vs. Chitosomes: Encapsulating Food Bioactives. Trends Food Sci. Technol. 2021, 108, 40–48. [Google Scholar] [CrossRef]
  134. Viegas, C.; Patrício, A.; Prata, J.; Nadhman, A.; Chintamaneni, P.; Fonte, P. Solid Lipid Nanoparticles vs. Nanostructured Lipid Carriers: A Comparative Review. Pharmaceutics 2023, 15, 1593. [Google Scholar] [CrossRef] [PubMed]
  135. Sotirova, Y. Solid Lipid Nanoparticles and Nanostructured Lipid Carriers: Current Perspectives in Wound Care. Scr. Sci. Pharm. 2023, 10, 24–34. [Google Scholar] [CrossRef]
  136. Akanda, M.; Mithu, M.S.H.; Douroumis, D. Solid Lipid Nanoparticles: An Effective Lipid-Based Technology for Cancer Treatment. J. Drug Deliv. Sci. Technol. 2023, 86, 104709. [Google Scholar] [CrossRef]
  137. Mall, J.; Naseem, N.; Haider, M.F.; Rahman, M.A.; Khan, S.; Siddiqui, S.N. Nanostructured Lipid Carriers as a Drug Delivery System: A Comprehensive Review with Therapeutic Applications. Intell. Pharm. 2024. [Google Scholar] [CrossRef]
  138. Souto, E.B.; Cano, A.; Martins-Gomes, C.; Coutinho, T.E.; Zielińska, A.; Silva, A.M. Microemulsions and Nanoemulsions in Skin Drug Delivery. Bioengineering 2022, 9, 158. [Google Scholar] [CrossRef]
  139. Musakhanian, J.; Osborne, D.W. Understanding Microemulsions and Nanoemulsions in (Trans)Dermal Delivery. AAPS PharmSciTech 2025, 26, 31. [Google Scholar] [CrossRef]
  140. Mohd-Nasir, H.; Abdul Aziz, Z.A.; Mohd Setapar, S.H. Chapter 14—In Vitro and in Vivo Safety Evaluation of Nanoemulsion as Skin Moisturizer. In Nanotechnology for the Preparation of Cosmetics Using Plant-Based Extracts; Mohd Setapar, S.H., Ahmad, A., Jawaid, M., Eds.; Elsevier: Amsterdam, The Netherlands, 2022; pp. 345–354. [Google Scholar]
  141. Burger, C.; Shahzad, Y.; Brummer, A.; Gerber, M.; du Plessis, J. Traversing the Skin Barrier with Nano-Emulsions. Curr. Drug Deliv. 2017, 14, 458–472. [Google Scholar] [CrossRef]
  142. Lopes, L. Overcoming the Cutaneous Barrier with Microemulsions. Pharmaceutics 2014, 6, 52–77. [Google Scholar] [CrossRef]
  143. Baranovskii, V.Y.; Ganev, V.G.; Petkova, V.B.; Voicheva, K.C.; Dimitrov, M.V. Hydrogels Based on Polycarboxylic Acid-Agar-Agar Complexes. Colloid. J. 2012, 74, 645–648. [Google Scholar] [CrossRef]
  144. Parhi, R.; Sahoo, S.K.; Das, A. Applications of Polysaccharides in Topical and Transdermal Drug Delivery: A Recent Update of Literature. Braz. J. Pharm. Sci. 2022, 58, e20802. [Google Scholar] [CrossRef]
  145. Dimitrov, M.; Dotcheva, D.; Lambov, N. Preparation and Characterization of Polyethylene Oxide Hydrogels with Cytisine. Acta Pharm. Turc. 2004, 46, 49–54. [Google Scholar]
  146. Singpanna, K.; Pornpitchanarong, C.; Patrojanasophon, P. Nanocomposite Patches for Transdermal Drug Delivery: A Review. Sci. Eng. Health Stud. 2024, 24010005. [Google Scholar] [CrossRef]
  147. Despotopoulou, D.; Lagopati, N.; Pispas, S.; Gazouli, M.; Demetzos, C.; Pippa, N. The Technology of Transdermal Delivery Nanosystems: From Design and Development to Preclinical Studies. Int. J. Pharm. 2022, 611, 121290. [Google Scholar] [CrossRef]
  148. Anitha, P.; Bhargavi, J.; Sravani, G.; Aruna, B.; Ramkanth, S. Recent Progress of Dendrimers in Drug Delivery for Cancer Therapy. Int. J. Appl. Pharm. 2018, 10, 34. [Google Scholar] [CrossRef]
  149. Nikzamir, M.; Hanifehpour, Y.; Akbarzadeh, A.; Panahi, Y. Applications of Dendrimers in Nanomedicine and Drug Delivery: A Review. J. Inorg. Organomet. Polym. Mater. 2021, 31, 2246–2261. [Google Scholar] [CrossRef]
  150. Pandita, D.; Madaan, K.; Kumar, S.; Poonia, N.; Lather, V. Dendrimers in Drug Delivery and Targeting: Drug-Dendrimer Interactions and Toxicity Issues. J. Pharm. Bioallied Sci. 2014, 6, 139. [Google Scholar] [CrossRef] [PubMed]
  151. Rout, S.; Bandaru, R.; Kenguva, G.; Hasan, N.; Alam, M.S.; Shukla, R.; Almalki, H.; Kesharwani, P.; Dandela, R. Dendrimers in Photodynamic Therapy. Nanomater. Photodyn. Ther. 2023, 281–305. [Google Scholar] [CrossRef]
  152. Kirkby, M.; Sabri, A.B.; Scurr, D.J.; Moss, G.P. Dendrimer-Mediated Permeation Enhancement of Chlorhexidine Digluconate: Determination of in Vitro Skin Permeability and Visualisation of Dermal Distribution. Eur. J. Pharm. Biopharm. 2021, 159, 77–87. [Google Scholar] [CrossRef] [PubMed]
  153. Kokaz, S.F.; Deb, P.K.; Borah, P.; Bania, R.; Venugopala, K.N.; Nair, A.B.; Singh, V.; Al-Shar’i, N.A.; Hourani, W.; Gupta, G.; et al. Dendrimers: Properties and Applications in Biomedical Field. Nanoeng. Biomater. 2021, 215–243. [Google Scholar] [CrossRef]
  154. Sorroza-Martínez, K.; Ruiu, A.; González-Méndez, I.; Rivera, E. Design and Properties of Dendrimers for Pharmaceutical Applications. Dendrimer-Based Nanother. 2021, 15–31. [Google Scholar] [CrossRef]
  155. Dangova, M.; Ivanova, N.; Andonova, V. Nanocarriers-Assisted Nose-to-Brain Delivery of Levodopa: Current Progress and Prospects. Appl. Sci. 2025, 15, 331. [Google Scholar] [CrossRef]
  156. Liang, R.; Wei, M.; Evans, D.G.; Duan, X. Inorganic Nanomaterials for Bioimaging, Targeted Drug Delivery and Therapeutics. ChemInform 2015, 46, 14071–14081. [Google Scholar] [CrossRef]
  157. Nasirzadeh, K.; Nazarian, S.; Gheibi Hayat, S.M. Inorganic Nanomaterials; A Brief Overview of the Applications and Developments in Sensing and Drug Delivery. J. Appl. Biotechnol. Rep. 2016, 3, 395–402. [Google Scholar]
  158. Mura, S.; Nicolas, J.; Couvreur, P. Stimuli-Responsive Nanocarriers for Drug Delivery. Nat. Mater. 2013, 12, 991–1003. [Google Scholar] [CrossRef]
  159. Pham, S.H.; Choi, Y.; Choi, J. Stimuli-Responsive Nanomaterials for Application in Antitumor Therapy and Drug Delivery. Pharmaceutics 2020, 12, 630. [Google Scholar] [CrossRef]
  160. Kang, M.-S.; Kwon, M.S.; Jang, H.J.; Jeong, S.J.; Han, D.-W.; Kim, K.S. Biosafety of Inorganic Nanomaterials for Theranostic Applications. Emerg. Mater. 2022, 5, 1995–2029. [Google Scholar] [CrossRef]
  161. Mishra, M.; Prasad, K.; Ramakrishn, S. Nanomaterials in Drug Delivery—Promises and Limitations. Nano Med Mater. 2023, 3, 38. [Google Scholar] [CrossRef]
  162. Thu, H.E.; Haider, M.A.; Khan, S.; Sohail, M.F.; Hussain, Z. Nanotoxicity Induced by Nanomaterials: A Review of Factors Affecting Nanotoxicity and Possible Adaptations. OpenNano 2023, 14, 100190. [Google Scholar] [CrossRef]
  163. Ahmad, M.Z.; Abdel-Wahab, B.A.; Alam, A.; Zafar, S.; Ahmad, J.; Ahmad, F.J.; Midoux, P.; Pichon, C.; Akhter, S. Toxicity of Inorganic Nanoparticles Used in Targeted Drug Delivery and Other Biomedical Application: An Updated Account on Concern of Biomedical Nanotoxicology. J. Nanosci. Nanotechnol. 2016, 16, 7873–7897. [Google Scholar] [CrossRef]
  164. Mahamuni-Badiger, P.P.; Dhanavade, M.J. Challenges and Toxicity Assessment of Inorganic Nanomaterials in Biomedical Applications: Current Status and Future Roadmaps. J. Drug Deliv. Sci. Technol. 2023, 87, 104806. [Google Scholar] [CrossRef]
  165. Tsikourkitoudi, V.; Henriques-Normark, B.; Sotiriou, G.A. Inorganic Nanoparticle Engineering against Bacterial Infections. Curr. Opin. Chem. Eng. 2022, 38, 100872. [Google Scholar] [CrossRef]
  166. Pei, Z.; Lei, H.; Cheng, L. Bioactive Inorganic Nanomaterials for Cancer Theranostics. Chem. Soc. Rev. 2023, 52, 2031–2081. [Google Scholar] [CrossRef] [PubMed]
  167. Pacyga, K.; Pacyga, P.; Szuba, E.; Viscardi, S.; Topola, E.; Duda-Madej, A. Nanotechnology Meets Phytotherapy: A Cutting-Edge Approach to Treat Bacterial Infections. Int. J. Mol. Sci. 2025, 26, 1254. [Google Scholar] [CrossRef] [PubMed]
  168. Ivanova, N.; Ermenlieva, N.; Simeonova, L.; Kolev, I.; Slavov, I.; Karashanova, D.; Andonova, V. Chlorhexidine–Silver Nanoparticle Conjugation Leading to Antimicrobial Synergism but Enhanced Cytotoxicity. Pharmaceutics 2023, 15, 2298. [Google Scholar] [CrossRef]
  169. Khan, S.S.; Ullah, I.; Ullah, S.; An, R.; Xu, H.; Nie, K.; Liu, C.; Liu, L. Recent Advances in the Surface Functionalization of Nanomaterials for Antimicrobial Applications. Materials 2021, 14, 6932. [Google Scholar] [CrossRef]
  170. Rizvi, M.; Gerengi, H.; Gupta, P. Functionalization of Nanomaterials: Synthesis and Characterization. Functionalized Nanomaterials for Corrosion Mitigation: Synthesis, Characterization, and Applications. Am. Chem. Soc. 2022, 1418, 1–26. [Google Scholar] [CrossRef]
  171. Gusmão, A.; de Albuquerque, P.B.S.; de Correia, A.C. Use of Metallic Nanoparticles Synthesized from Plant Extracts in Wound Healing—A Review. Appl. Nano 2024, 5, 205–226. [Google Scholar] [CrossRef]
  172. Shanmugasundaram, T.; Shanmugasundaram, T.; Radhakrishnan, M.; Gopikrishnan, V.; Kadirvelu, K.; Balagurunathan, R. In Vitro Antimicrobial and in Vivo Wound Healing Effect of Actinobacterially Synthesised Nanoparticles of Silver, Gold and Their Alloy. RSC Adv. 2017, 7, 51729–51743. [Google Scholar] [CrossRef]
  173. Jangid, H.; Singh, S.; Kashyap, P.K.; Singh, A. Advancing Biomedical Applications: An in-Depth Analysis of Silver Nanoparticles in Antimicrobial, Anticancer, and Wound Healing Roles. Front. Pharmacol. 2024, 15, 1438227. [Google Scholar] [CrossRef]
  174. Clichici, S.; Filip, A. In Vivo Assessment of Nanomaterials Toxicity. In Nanomaterials; Soloneski, S., Larramendy, M.L., Eds.; IntechOpen: Rijeka, Croatia, 2015; pp. 93–121. [Google Scholar]
  175. Dhanalekshmi, K.I.; Sangeetha, K.; Magesan, P.; Johnson, J.; Zhang, X.; Jayamoorthy, K. Photodynamic Cancer Therapy: Role of Ag- and Au-Based Hybrid Nano-Photosensitizers. J. Biomol. Struct. Dyn. 2020, 40, 4766–4773. [Google Scholar] [CrossRef]
  176. Augustine, R.; Hasan, A.; Primavera, R.; Wilson, R.J.; Thakor, A.S.; Kevadiya, B.D. Cellular Uptake and Retention of Nanoparticles: Insights on Particle Properties and Interaction with Cellular Components. Mater. Today Commun. 2020, 25, 101692. [Google Scholar] [CrossRef]
  177. Wang, X. The Comparison of Titanium Dioxide and Zinc Oxide Used in Sunscreen Based on Their Enhanced Absorption. Appl. Comput. Eng. 2023, 24, 237–245. [Google Scholar] [CrossRef]
  178. Wu, H. The Application of Metal Oxide Nanoparticles in Sunscreen and Their Potential Risks. Appl. Comput. Eng. 2023, 24, 210–215. [Google Scholar] [CrossRef]
  179. Vujovic, M.; Kostic, E. Titanium Dioxide and Zinc Oxide Nanoparticles in Sunscreens: A Review of Toxicological Data. J. Cosmet. Sci. 2019, 70, 223–234. [Google Scholar]
  180. Popov, A.; Zhao, X.; Zvyagin, A.; Lademann, J.; Roberts, M.; Sanchez, W.; Priezzhev, A.; Myllylä, R. ZnO and TiO2 Particles: A Study on Nanosafety and Photoprotection. Biophotonics Photonic Solut. Better Health Care II 2010, 7715, 77153G. [Google Scholar] [CrossRef]
  181. Osmond, M.J.; Mccall, M.J. Zinc Oxide Nanoparticles in Modern Sunscreens: An Analysis of Potential Exposure and Hazard. Nanotoxicology 2010, 4, 15–41. [Google Scholar] [CrossRef]
  182. Gulson, B.; McCall, M.J.; Bowman, D.M.; Pinheiro, T. A Review of Critical Factors for Assessing the Dermal Absorption of Metal Oxide Nanoparticles from Sunscreens Applied to Humans, and a Research Strategy to Address Current Deficiencies. Arch. Toxicol. 2015, 89, 1909–1930. [Google Scholar] [CrossRef]
  183. Warheit, D.B. Safety of titanium dioxide (E171) as a food additive for humans. Front. Toxicol. 2024, 19, 1333746. [Google Scholar] [CrossRef] [PubMed]
  184. Goette, D.K. Topical Chemotherapy with 5-Fluorouracil: A Review. J. Am. Acad. Dermatol. 1981, 4, 633–649. [Google Scholar] [CrossRef]
  185. du Vivier, A. Topical Cytostatic Drugs in the Treatment of Skin Cancer. Clin. Exp. Dermatol. 1982, 7, 89–92. [Google Scholar] [CrossRef]
  186. Kim, S.; Woo, Y.R.; Cho, S.H.; Lee, J.D.; Kim, H.S. Clinical Efficacy of 5-Fluorouracil and Bleomycin in Dermatology. J. Clin. Med. 2024, 13, 335. [Google Scholar] [CrossRef] [PubMed]
  187. Available online: https://www.drugs.com/monograph/fluorouracil-topical.html (accessed on 4 April 2025).
  188. Bargman, H.; Hochman, J. Topical treatment of Bowen’s disease with 5-Fluorouracil. J. Cutan. Med. Surg. 2003, 7, 101–105. [Google Scholar] [CrossRef]
  189. Salim, A.; Leman, J.; McColl, J.; Chapman, R.; Morton, C. Randomized comparison of photodynamic therapy with topical 5-fluorouracil in Bowen’s disease. Br. J. Dermatol. 2003, 148, 539–543. [Google Scholar] [CrossRef] [PubMed]
  190. Ryan, R.; Krementz, E.; Litwin, M. A Role for Topical 5-Fluorouracil Therapy in Melanoma. J. Surg. Oncol. 1988, 38, 250–256. [Google Scholar] [CrossRef] [PubMed]
  191. Metterle, L.; Nelson, C.; Patel, N. Intralesional 5-fluorouracil (FU) as a treatment for nonmelanoma skin cancer (NMSC): A review. J. Am. Acad. Dermatol. 2016, 74, 552–557. [Google Scholar] [CrossRef]
  192. Maxfield, L.; Shah, M.; Schwartz, C.; Tanner, L.S.; Appel, J. Intralesional 5-fluorouracil for the treatment of squamous cell carcinomas. J. Am. Acad. Dermatol. 2021, 84, 1696–1697. [Google Scholar] [CrossRef]
  193. Luu, W.; McRae, M.Y. Intralesional 5-fluorouracil as a management for cutaneous squamous cell carcinomas: A rural Australian retrospective case series. Australas J. Dermatol. 2023, 64, 556–559. [Google Scholar] [CrossRef]
  194. Morse, L.G.; Kendrick, C.; Hooper, D.; Ward, H.; Parry, E. Treatment of squamous cell carcinoma with intralesional 5-Fluorouracil. Dermatol. Surg. 2003, 29, 1150–1153. [Google Scholar]
  195. Leonard, A.L.; Hanke, C.W. Treatment of giant keratoacanthoma with intralesional 5-fluorouracil. J. Drugs Dermatol. 2006, 5, 454–456. [Google Scholar]
  196. Peng, L.; Zhang, X.; Du, B.; Sun, L.-J.; Zhao, Y. Mechanism of 5-Fluorouracil Nanoparticles on Releasing Skin Squamous Cell Carcinoma through Regulation of Wnt/β-Catenin Expression. Mater. Express 2022, 12, 220–226. [Google Scholar] [CrossRef]
  197. Tian, J.; Tian, J.; Zhang, D.; Kurbatov, V.; Kurbatov, V.; Wang, Q.; Wang, Y.; Fang, D.; Wu, L.; Bosenberg, M.; et al. 5-Fluorouracil Efficacy Requires Anti-Tumor Immunity Triggered by Cancer-Cell-Intrinsic STING. EMBO J. 2021, 40, e106065. [Google Scholar] [CrossRef]
  198. Dai, X.-L.; Voronin, A.P.; Gao, W.; Perlovich, G.L.; Lu, T.-B.; Chen, J.-M. Intermolecular Interactions and Permeability of 5-Fluorouracil Cocrystals with a Series of Isomeric Hydroxybenzoic Acids: A Combined Theoretical and Experimental Study. CrystEngComm 2019, 21, 5095–5105. [Google Scholar] [CrossRef]
  199. Nikam, A.N.; Jacob, A.; Raychaudhuri, R.; Fernandes, G.; Pandey, A.; Rao, V.; Ahmad, S.F.; Pannala, A.S.; Mutalik, S. Topical Micro-Emulsion of 5-Fluorouracil by a Twin Screw Processor-Based Novel Continuous Manufacturing Process for the Treatment of Skin Cancer: Preparation and In Vitro and In Vivo Evaluations. Pharmaceutics 2023, 15, 2175. [Google Scholar] [CrossRef]
  200. Available online: https://pubmed.ncbi.nlm.nih.gov/?term=5-fluorouracil+skin+cancer&filter=datesearch.y_1 (accessed on 4 April 2025).
  201. Siddalingam, R.; Chidambaram, K. Topical Nano-Delivery of 5-Fluorouracil: Preparation and Characterization of Water-in-Oil Nanoemulsion. Trop. J. Pharm. Res. 2016, 15, 2311–2319. [Google Scholar] [CrossRef]
  202. Nawaz, A.; Latif, M.S.; Alnuwaiser, M.A.; Ullah, S.; Iqbal, M.; Alfatama, M.; Lim, V. Synthesis and Characterization of Chitosan-Decorated Nanoemulsion Gel of 5-Fluorouracil for Topical Delivery. Gels 2022, 8, 412. [Google Scholar] [CrossRef]
  203. Crisóstomo, L.C.; Carvalho, G.S.G.; Leal, L.K.A.M.; de Araújo, T.G.; Nogueira, K.A.B.; da Silva, D.A.; Ribeiro, F.d.O.S.; Petrilli, R.; Eloy, J.O. Sorbitan Monolaurate–Containing Liposomes Enhance Skin Cancer Cell Cytotoxicity and in Association with Microneedling Increase the Skin Penetration of 5-Fluorouracil. AAPS Pharmscitech 2022, 23, 212. [Google Scholar] [CrossRef] [PubMed]
  204. Calienni, M.N.; Prieto, M.J.; Couto, V.M.; de Paula, E.; del Alonso, S.V.; Montanari, J. 5-Fluorouracil-Loaded Ultradeformable Liposomes for Skin Therapy. AIP Conf. Proc. 2018, 1990, 020024. [Google Scholar] [CrossRef]
  205. Cosco, D.; Paolino, D.; Maiuolo, J.; Marzio, L.D.; Carafa, M.; Ventura, C.A.; Fresta, M. Ultradeformable Liposomes as Multidrug Carrier of Resveratrol and 5-Fluorouracil for Their Topical Delivery. Int. J. Pharm. 2015, 489, 1–10. [Google Scholar] [CrossRef] [PubMed]
  206. Petrilli, R.; Eloy, J.O.; Saggioro, F.P.; Chesca, D.L.; de Souza, M.C.; Dias, M.V.S.; daSilva, L.L.P.; Lee, R.J.; Lopez, R.F.V. Skin Cancer Treatment Effectiveness Is Improved by Iontophoresis of EGFR-Targeted Liposomes Containing 5-FU Compared with Subcutaneous Injection. J. Control. Release 2018, 283, 151–162. [Google Scholar] [CrossRef] [PubMed]
  207. Yaman, Ü.; Aslan, M.; Ozturk, S.; Ulubayram, K.; Eroğlu, İ. Surface Modified Nanoliposome Formulations Provide Sustained Release for 5-FU and Increase Cytotoxicity on A431 Cell Line. Pharm. Dev. Technol. 2020, 25, 1192–1203. [Google Scholar] [CrossRef]
  208. Khalid, W.; Shah, K.U.; Saeed, M.D.; Nawaz, A.; Rehman, F.U.; Shoaib, M.; Rehman, M.U.; Alasmari, A.; Alharbi, M.; Alasmari, F. 5-Fluorouracil-Loaded Hyaluronic Acid-Coated Niosomal Vesicles: Fabrication and Ex Vivo Evaluation for Skin Drug Delivery. ACS Omega 2023, 8, 45405–45413. [Google Scholar] [CrossRef] [PubMed]
  209. Khan, N.R.; Wong, T.W. 5-Fluorouracil Ethosomes-Skin Deposition and Melanoma Permeation Synergism with Microwave. Artif. Cells Nanomed. Biotechnol. 2018, 46, 568–577. [Google Scholar] [CrossRef] [PubMed]
  210. Puri, R.; Jain, S. Ethogel Topical Formulation for Increasing the Local Bioavailability of 5-Fluorouracil: A Mechanistic Study. Anti-Cancer Drugs 2012, 23, 923–934. [Google Scholar] [CrossRef]
  211. Hasan, N.; Imran, M.; Sheikh, A.; Tiwari, N.; Jaimini, A.; Kesharwani, P.; Jain, G.; Ahmad, F.J. Advanced Multifunctional Nano-Lipid Carrier Loaded Gel for Targeted Delivery of 5-Flurouracil and Cannabidiol against Non-Melanoma Skin Cancer. Environ. Res. 2023, 233, 116454. [Google Scholar] [CrossRef]
  212. Rajinikanth, P.S.; Chellian, J. Development and Evaluation of Nanostructured Lipid Carrier-Based Hydrogel for Topical Delivery of 5-Fluorouracil. Int. J. Nanomed. 2016, 11, 5067–5077. [Google Scholar] [CrossRef]
  213. Ali, A.; Madni, A.; Shah, H.A.; Jamshaid, T.; Jan, N.; Khan, S.; Khan, M.M.; Mahmood, M.A. Solid Lipid-Based Nanoparticulate System for Sustained Release and Enhanced in-Vitro Cytotoxic Effect of 5-Fluorouracil on Skin Melanoma and Squamous Cell Carcinoma. PLoS ONE 2023, 18, e0281004. [Google Scholar] [CrossRef]
  214. 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]
  215. Shenoy, V.S.; Gude, R.P.; Murthy, R.S.R. In Vitro Anticancer Evaluation of 5-Fluorouracil Lipid Nanoparticles Using B16F10 Melanoma Cell Lines. Int. Nano Lett. 2013, 3, 36. [Google Scholar] [CrossRef]
  216. Patel, G.; Yadav, B.K.N. Study of 5-Fluorouracil Loaded Chitosan Nanoparticles for Treatment of Skin Cancer. Recent Pat. Nanotechnol. 2020, 14, 210–224. [Google Scholar] [CrossRef] [PubMed]
  217. Patel, G.; Yadav, B.K.N. Formulation, Characterization and In Vitro Cytotoxicity of 5-Fluorouracil Loaded Polymeric Electrospun Nanofibers for the Treatment of Skin Cancer. Recent Pat. Nanotechnol. 2019, 13, 114–128. [Google Scholar] [CrossRef]
  218. Skok, K.; Zidarič, T.; Orthaber, K.; Pristovnik, M.; Kostevšek, N.; Žužek Rožman, K.; Šturm, S.; Gradišnik, L.; Maver, U.; Maver, T. Novel Methacrylate-Based Multilayer Nanofilms with Incorporated FePt-Based Nanoparticles and the Anticancer Drug 5-Fluorouracil for Skin Cancer Treatment. Pharmaceutics 2022, 14, 689. [Google Scholar] [CrossRef]
  219. Safwat, M.A.; Safwat, M.A.; Soliman, G.M.; Soliman, G.M.; Sayed, D.; Attia, M.A. Fluorouracil-Loaded Gold Nanoparticles for the Treatment of Skin Cancer: Development, in Vitro Characterization, and in Vivo Evaluation in a Mouse Skin Cancer Xenograft Model. Mol. Pharm. 2018, 15, 2194–2205. [Google Scholar] [CrossRef]
  220. Pourmanouchehri, Z.; Ebrahimi, S.; Limoee, M.; Jalilian, F.; Janfaza, S.; Vosoughi, A.; Behbood, L. Controlled Release of 5-Fluorouracil to Melanoma Cells Using a Hydrogel/Micelle Composites Based on Deoxycholic Acid and Carboxymethyl Chitosan. Int. J. Biol. Macromol. 2022, 206, 159–166. [Google Scholar] [CrossRef] [PubMed]
  221. Mohammad, O.; Faisal, S.M.; Ahmad, N. Bio-Mediated Synthesis of 5-FU Based Nanoparticles Employing Orange Fruit Juice: A Novel Drug Delivery System to Treat Skin Fibrosarcoma in Model Animals. Sci. Rep. 2019, 9, 12288. [Google Scholar] [CrossRef] [PubMed]
  222. Shojaei, S.; Doostan, M.; Mohammadi Motlagh, H.; Esnaashari, S.; Maleki, H. Development of 5-Fluorouracil/Etoposide Co-Loaded Electrospun Nanofibrous Scaffold for Localized Anti-Melanoma Therapy. J. Appl. Biomater. Funct. Mater. 2024, 22, 22808000241284439. [Google Scholar] [CrossRef]
  223. Nanda, J.; Bermudez, R. Imiquimod. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  224. Garcia-Mouronte, E.; Berná-Rico, E.; de Nicolas-Ruanes, B.; Azcarraga-Llobet, C.; Bea-Ardebol, S. Imiquimod as Local Immunotherapy in the Management of Premalignant Cutaneous Conditions and Skin Cancer. Int. J. Mol. Sci. 2023, 24, 10835. [Google Scholar] [CrossRef] [PubMed]
  225. Wolff, F.; Loipetzberger, A.; Gruber, W.; Esterbauer, H.; Aberger, F.; Frischauf, A. Imiquimod Directly Inhibits Hedgehog Signalling by Stimulating Adenosine Receptor/Protein Kinase A-Mediated GLI Phosphorylation. Oncogene 2013, 32, 5574–5581. [Google Scholar] [CrossRef]
  226. Vidal, D.; Matias-Guiu, X.; Alomar, A. Open Study of the Efficacy and Mechanism of Action of Topical Imiquimod in Basal Cell Carcinoma. Clin. Exp. Dermatol. 2004, 29, 518–525. [Google Scholar] [CrossRef]
  227. Yokogawa, M.; Takaishi, M.; Nakajima, K.; Kamijima, R.; DiGiovanni, J.; Sano, S. Imiquimod Attenuates the Growth of UVB-Induced SCC in Mice through Th1/Th17 Cells. Mol. Carcinog. 2013, 52, 760–769. [Google Scholar] [CrossRef]
  228. Fan, Q.; Cohen, S.; John, B.; Riker, A. Melanoma in Situ Treated with Topical Imiquimod for Management of Persistently Positive Margins: A Review of Treatment Methods. Ochsner J. 2015, 15, 443–447. [Google Scholar]
  229. Ray, C.; Kluk, M.; Grin, C.; Grant-Kels, J. Successful Treatment of Malignant Melanoma in Situ with Topical 5% Imiquimod Cream. Int. J. Dermatol. 2005, 44, 428–434. [Google Scholar] [CrossRef]
  230. Patel, G.; Goodwin, R.; Chawla, M.; Laidler, P.; Price, P.; Finlay, A.; Motley, R. Imiquimod 5% Cream Monotherapy for Cutaneous Squamous Cell Carcinoma in Situ (Bowen’s Disease): A Randomized, Double-Blind, Placebo-Controlled Trial. J. Am. Acad. Dermatol. 2006, 54, 1025–1032. [Google Scholar] [CrossRef]
  231. Casadei, B.R.; Lotierzo, M.C.G.; Malheiros, B.; Barbosa, L.R.S. Drug Repurposing and Nanoparticles: New Strategies against Leishmaniasis. In Applications of Nanobiotechnology for Neglected Tropical Diseases; Academic Press: Cambridge, MA, USA, 2021; pp. 217–241. [Google Scholar]
  232. Berman, B. Safety, Efficacy, and Patient Acceptability of Imiquimod for Topical Treatment of Actinic Keratoses. Clin. Cosmet. Investig. Dermatol. 2011, 4, 35–40. [Google Scholar] [CrossRef] [PubMed]
  233. Sorgi, D.; Sartori, A.; Germani, S.; Gentile, R.N.; Bianchera, A.; Bettini, R. Imiquimod Solubility in Different Solvents: An Interpretative Approach. Pharmaceutics 2024, 16, 282. [Google Scholar] [CrossRef] [PubMed]
  234. Kim, S.; Abdella, S.; Abid, F.; Afinjuomo, F.; Youssef, S.; Holmes, A.; Song, Y.; Vaidya, S.; Garg, S. Development and Optimization of Imiquimod-Loaded Nanostructured Lipid Carriers Using a Hybrid Design of Experiments Approach. Int. J. Nanomed. 2023, 18, 1007–1029. [Google Scholar] [CrossRef] [PubMed]
  235. Al-Mayahy, M.H.; Marlow, M.; Scurr, D.J. The Complementary Role of ToF-SIMS in the Assessment of Imiquimod Permeated into the Skin from a Microemulsion Dosage Form. Al Mustansiriyah J. Pharm. Sci. 2019, 19, 196–210. [Google Scholar] [CrossRef]
  236. Ghezzi, M.; Pescina, S.; Delledonne, A.; Ferraboschi, I.; Sissa, C.; Terenziani, F.; Remiro, P.D.F.R.; Santi, P.; Nicoli, S. Improvement of Imiquimod Solubilization and Skin Retention via TPGS Micelles: Exploiting the Co-Solubilizing Effect of Oleic Acid. Pharmaceutics 2021, 13, 1476. [Google Scholar] [CrossRef]
  237. De Paula, D.; Martins, C.A.; Bentley, M.V.L.B. Development and Validation of HPLC Method for Imiquimod Determination in Skin Penetration Studies. Biomed. Chromatogr. 2008, 22, 1416–1423. [Google Scholar] [CrossRef]
  238. Layek, B.; Rahman Nirzhor, S.S.; Rathi, S.; Kandimalla, K.K.; Wiedmann, T.S.; Prabha, S. Design, Development, and Characterization of Imiquimod-Loaded Chitosan Films for Topical Delivery. AAPS PharmSciTech 2019, 20, 58. [Google Scholar] [CrossRef]
  239. Panoutsopoulou, E.; Zbytovská, J.; Vávrová, K.; Paraskevopoulos, G. Phospholipid-Based Microemulsions for Cutaneous Imiquimod Delivery. Pharmaceuticals 2022, 15, 515. [Google Scholar] [CrossRef]
  240. Telò, I.; Del Favero, E.; Cantù, L.; Frattini, N.; Pescina, S.; Padula, C.; Santi, P.; Sonvico, F.; Nicoli, S. Gel-like TPGS-Based Microemulsions for Imiquimod Dermal Delivery: Role of Mesostructure on the Uptake and Distribution into the Skin. Mol. Pharm. 2017, 14, 3281–3289. [Google Scholar] [CrossRef]
  241. Jadhav, S.T.; Salunkhe, V.R.; Bhinge, S.D. Nanoemulsion Drug Delivery System Loaded with Imiquimod: A QbD-Based Strategy for Augmenting Anti-Cancer Effects. Future J. Pharm. Sci. 2023, 9, 120. [Google Scholar] [CrossRef]
  242. Petrová, E.; Chvíla, S.; Balouch, M.; Štěpánek, F.; Zbytovská, J. Nanoformulations for Dermal Delivery of Imiquimod: The Race of “Soft” against “Hard”. Int. J. Pharm. 2023, 648, 123577. [Google Scholar] [CrossRef] [PubMed]
  243. Caimi, A.; Ramirez, C.; Perez, A.; Romero, E.; Morilla, M. In Vitro Anti-Melanoma Activity of Imiquimod in Ultradeformable Nanovesicles. Drug Dev. Ind. Pharm. 2022, 48, 657–666. [Google Scholar] [CrossRef] [PubMed]
  244. Tampucci, S.; Guazzelli, L.; Burgalassi, S.; Carpi, S.; Carpi, S.; Chetoni, P.; Mezzetta, A.; Nieri, P.; Polini, B.; Pomelli, C.S.; et al. pH-Responsive Nanostructures Based on Surface Active Fatty Acid-Protic Ionic Liquids for Imiquimod Delivery in Skin Cancer Topical Therapy. Pharmaceutics 2020, 12, 1078. [Google Scholar] [CrossRef]
  245. Ma, M.; Wang, J.; Guo, F.; Lei, M.; Tan, F.; Li, N. Development of Nanovesicular Systems for Dermal Imiquimod Delivery: Physicochemical Characterization and in Vitro/in Vivo Evaluation. J. Mater. Sci. Mater. Med. 2015, 26, 192. [Google Scholar] [CrossRef]
  246. Carreño, G.F.; Alvarez-Figueroa, M.J.; González-Aramundiz, J.V. Dextran Nanocapsules with ω-3 in Their Nucleus: An Innovative Nanosystem for Imiquimod Transdermal Delivery. Pharmaceutics 2022, 14, 2445. [Google Scholar] [CrossRef]
  247. Argenziano, M.; Haimhoffer, Á.; Bastiancich, C.; Jicsinszky, L.; Caldera, F.; Trotta, F.; Scutera, S.; Alotto, D.; Fumagalli, M.; Musso, T.; et al. In Vitro Enhanced Skin Permeation and Retention of Imiquimod Loaded in β-Cyclodextrin Nanosponge Hydrogel. Pharmaceutics 2019, 11, 138. [Google Scholar] [CrossRef]
  248. 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, 195. [Google Scholar] [CrossRef] [PubMed]
  249. Dao, D.-P.D.; Sahni, V.N.; Sahni, D.R.; Balogh, E.A.; Grada, A.; Feldman, S.R. 1% Tirbanibulin Ointment for the Treatment of Actinic Keratoses. Ann. Pharmacother. 2021, 56, 494–500. [Google Scholar] [CrossRef]
  250. Schlesinger, T.E.; Stockfleth, E.; Grada, A.; Berman, B. Tirbanibulin for Actinic Keratosis: Insights into the Mechanism of Action. Clin. Cosmet. Investig. Dermatol. 2022, 15, 2495–2506. [Google Scholar] [CrossRef]
  251. Sajkiewicz, I.; Miga-Orczykowska, N.; Lemieszek, P.; Jasiuk, I.; Pustelniak, M.; Wójtowicz, J.; Krukar, K.; Rudnicka, K.; Łukaszewska, E.; Kister, K. Tirbanibulin as a Novel Treatment in Actinic Keratosis: A Literature Review. J. Educ. Health Sport. 2024, 70, 55328. [Google Scholar] [CrossRef]
  252. Atzeni, F.; Masala, I.; Sarzi-Puttini, P. A Review of Chronic Musculoskeletal Pain: Central and Peripheral Effects of Diclofenac. Pain Ther. 2018, 7, 163–177. [Google Scholar] [CrossRef]
  253. Müller-Decker, K. Cyclooxygenase-Dependent Signaling Is Causally Linked to Non-Melanoma Skin Carcinogenesis: Pharmacological, Genetic, and Clinical Evidence. Cancer Metastasis Rev. 2011, 30, 343–361. [Google Scholar] [CrossRef]
  254. Zhan, H.; Zheng, H. The Role of Topical Cyclo-Oxygenase-2 Inhibitors in Skin Cancer: Treatment and Prevention. Am. J. Clin. Dermatol. 2007, 8, 195–200. [Google Scholar] [CrossRef]
  255. Rodust, P.M.; Fecker, L.F.; Stockfleth, E.; Eberle, J. Activation of Mitochondrial Apoptosis Pathways in Cutaneous Squamous Cell Carcinoma Cells by Diclofenac/Hyaluronic Acid Is Related to Upregulation of Bad as Well as Downregulation of Mcl-1 and Bcl-w. Exp. Dermatol. 2012, 21, 520–525. [Google Scholar] [CrossRef] [PubMed]
  256. Brinkhuizen, T.; Frencken, K.J.A.; Nelemans, P.J.; Hoff, M.L.S.; Kelleners-Smeets, N.W.J.; zur Hausen, A.; van der Horst, M.P.J.; Rennspiess, D.; Winnepenninckx, V.; van Steensel, M.A.M.; et al. The Effect of Topical Diclofenac 3% and Calcitriol 3 Μg/g on Superficial Basal Cell Carcinoma (sBCC) and Nodular Basal Cell Carcinoma (nBCC): A Phase II, Randomized Controlled Trial. J. Am. Acad. Dermatol. 2016, 75, 126–134. [Google Scholar] [CrossRef]
  257. Jarvis, B.; Figgitt, D. Topical 3% Diclofenac in 2.5% Hyaluronic Acid Gel: A Review of Its Use in Patients with Actinic Keratoses. Am. J. Clin. Dermatol. 2003, 4, 203–213. [Google Scholar] [CrossRef] [PubMed]
  258. Pommergaard, H.-C.; Burcharth, J.; Rosenberg, J.; Raskov, H. Topical Combination of Diclofenac, Calcipotriol, and Difluoromethylornithine Has Beneficial Effects Comparable to 5-Fluorouracil for the Treatment of Non-Melanoma Skin Cancer in Mice. J. Chemother. 2014, 26, 105–110. [Google Scholar] [CrossRef]
  259. El-Khalawany, M.; Saudi, W.M.; Ahmed, E.Z.; Mosbeh, A.-S.; Sameh, A.; Rageh, M.A. The Combined Effect of CO2 Laser, Topical Diclofenac 3%, and Imiquimod 5% in Treating High-Risk Basal Cell Carcinoma. J. Cosmet. Dermatol. 2021, 21, 2049–2055. [Google Scholar] [CrossRef] [PubMed]
  260. Lucena, S.R.; Salazar, N.; Gracia-Cazaña, T.; Zamarrón, A.; González, S.; González, S.; Juarranz, A.; Gilaberte, Y. Combined Treatments with Photodynamic Therapy for Non-Melanoma Skin Cancer. Int. J. Mol. Sci. 2015, 16, 25912–25933. [Google Scholar] [CrossRef]
  261. Tampucci, S.; Carpi, S.; Digiacomo, M.; Polini, B.; Fogli, S.; Burgalassi, S.; Macchia, M.; Nieri, P.; Manera, C.; Monti, D. Diclofenac-Derived Hybrids for Treatment of Actinic Keratosis and Squamous Cell Carcinoma. Molecules 2019, 24, 1793. [Google Scholar] [CrossRef]
  262. Fidler, B.; Goldberg, T. Ingenol Mebutate Gel (Picato): A Novel Agent for the Treatment of Actinic Keratoses. Pharm. Ther. 2014, 39, 40–46. [Google Scholar]
  263. Gras, J. Ingenol Mebutate: A New Option for Actinic Keratosis Treatment. Drugs Today 2013, 49, 15. [Google Scholar] [CrossRef]
  264. Freiberger, S.N.; Cheng, P.F.; Iotzova-Weiss, G.; Neu, J.; Liu, Q.; Dziunycz, P.; Zibert, J.R.; Dummer, R.; Skak, K.; Levesque, M.P.; et al. Data from Ingenol Mebutate Signals via PKC/MEK/ERK in Keratinocytes and Induces Interleukin Decoy Receptors IL1R2 and IL13RA2. Mol. Cancer Ther. 2023, 14, 2132. [Google Scholar] [CrossRef] [PubMed]
  265. Challacombe, J.; Suhrbier, A.; Parsons, P. Neutrophils Are a Key Component of the Antitumor Efficacy of Topical Chemotherapy with Ingenol-3-Angelate. J. Immunol. 2006, 177, 8123–8132. [Google Scholar] [CrossRef]
  266. Velin, M.; Cardot-Leccia, N.; Cathelineau, A.C.; Duteil, L.; Queille-Roussel, C.; Passeron, T.; Bahadoran, P. Efficacy and Safety of 0.05% Ingenol Mebutate in the Treatment of Basal Cell Carcinoma: A Prospective Study. Ski. Health Dis. 2022, 3, e150. [Google Scholar] [CrossRef] [PubMed]
  267. Banzhaf, C.A.; Phothong, W.; Suku, M.H.; Ulrich, M.; Philipsen, P.A.; Mogensen, M.; Haedersdal, M. Basal Cell Carcinoma Treated with Combined Ablative Fractional Laser and Ingenol Mebutate—An Exploratory Study Monitored by Optical Coherence Tomography and Reflectance Confocal Microscopy. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 502–509. [Google Scholar] [CrossRef] [PubMed]
  268. Tan, I.J.; Pathak, G.; Silver, F.H. Topical Treatments for Basal Cell Carcinoma and Actinic Keratosis in the United States. Cancers 2023, 15, 3927. [Google Scholar] [CrossRef]
  269. An, J.H.; Shin, J.U.; Kim, H.J.; Lee, H.J.; Yoon, M.S.; Kim, D.-H. Comparison of the Treatment Outcomes of Photodynamic Therapy and Ingenol Mebutate in Bowen’s Disease: A Retrospective Observational Study. Ann. Dermatol. 2020, 32, 47–52. [Google Scholar] [CrossRef]
  270. Gupta, A.K.; Paquet, M. Mebutate: A Promising Treatment for Actinic Keratoses and Nonmelanoma Skin Cancers. J. Cutan. Med. Surgery 2013, 17, 173–179. [Google Scholar] [CrossRef]
  271. Alkhalaf, A.; Hofbauer, G.F.L. Ingenol Mebutate 150 Mg as Physician-Directed Treatment of Bowen’s Disease Under Occlusion. Dermatology 2016, 232, 17–19. [Google Scholar] [CrossRef] [PubMed]
  272. Jedlowski, P. Ingenol Mebutate Is Associated With Increased Reporting Odds for Squamous Cell Carcinoma in Actinic Keratosis Patients, a Pharmacovigilance Study of the FDA Adverse Event Reporting System (FAERS). J. Cutan. Med. Surg. 2022, 27, 39–43. [Google Scholar] [CrossRef]
  273. Wang, Y.; Wang, Y.; Lipner, S.R.; Lipner, S.R. Retrospective Analysis of Squamous Cell Carcinoma Associated with Ingenol Mebutate Reported to the US Food and Drug Administration. Dermatol. Ther. 2020, 33, e14114. [Google Scholar] [CrossRef]
  274. Ramsay, J.; Suhrbier, A.; Aylward, J.; Ogbourne, S.; Cozzi, S.; Poulsen, M.; Baumann, K.; Welburn, P.; Redlich, G.; Parsons, P. The Sap from Euphorbia Peplus Is Effective against Human Nonmelanoma Skin Cancers. Br. J. Dermatol. 2011, 164, 633–636. [Google Scholar] [CrossRef]
  275. Jørgensen, L.; McKerrall, S.J.; Kuttruff, C.A.; Ungeheuer, F.; Felding, J.; Baran, P.S. 14-Step Synthesis of (+)-Ingenol from (+)-3-Carene. Science 2013, 341, 878–882. [Google Scholar] [CrossRef]
  276. Tang, H.; Yusuff, N.; Wood, J.L. Progress Toward the Total Synthesis of Ingenol: Construction of the Complete Carbocyclic Skeleton. ChemInform 2001, 32. [Google Scholar] [CrossRef]
  277. Liang, X.; Grue-Sørensen, G.; Månsson, K.; Vedsø, P.; Soor, A.; Stahlhut, M.; Bertelsen, M.; Engell, K.M.; Högberg, T. Syntheses, Biological Evaluation and SAR of Ingenol Mebutate Analogues for Treatment of Actinic Keratosis and Non-Melanoma Skin Cancer. Bioorganic Med. Chem. Lett. 2013, 23, 5624–5629. [Google Scholar] [CrossRef]
  278. Bertelsen, M.; Stahlhut, M.; Grue-Sørensen, G.; Liang, X.; Christensen, G.B.; Skak, K.; Engell, K.M.; Högberg, T. Ingenol Disoxate: A Novel 4-Isoxazolecarboxylate Ester of Ingenol with Improved Properties for Treatment of Actinic Keratosis and Other Non-Melanoma Skin Cancers. Dermatol. Ther. 2016, 6, 599–626. [Google Scholar] [CrossRef] [PubMed]
  279. Trémezaygues, L.; Reichrath, J. Vitamin D Analogs in the Treatment of Psoriasis: Where Are We Standing and Where Will We Be Going? Dermatoendocrinology 2011, 3, 180–186. [Google Scholar] [CrossRef]
  280. Chakraborty, D.; Aggarwal, K. Comparative Evaluation of Efficacy and Safety of Calcipotriol versus Calcitriol Ointment, Both in Combination with Narrow-Band Ultraviolet B Phototherapy in the Treatment of Stable Plaque Psoriasis. Photodermatol. Photoimmunol. Photomed. 2023, 39, 512–519. [Google Scholar] [CrossRef]
  281. Rosenberg, A.; Tabacchi, M.; Ngo, K. Skin Cancer Precursor Immunotherapy for Squamous Cell Carcinoma Prevention. JCI Insight 2019, 4, e125476. [Google Scholar] [CrossRef]
  282. Azin, M.; Mahon, A.; Isaacman, S. Topical Calcipotriol plus 5-Fluorouracil Immunotherapy for Actinic Keratosis Treatment. JID Innov. 2022, 2, 100104. [Google Scholar] [CrossRef]
  283. Cunningham, T.; Tabacchi, M.; Eliane, J. Randomized Trial of Calcipotriol Combined with 5-Fluorouracil for Skin Cancer Precursor Immunotherapy. J. Clin. Investig. 2017, 127, 106–116. [Google Scholar] [CrossRef] [PubMed]
  284. Moore, A.; Nguyen, M.; Moore, S. Cyclic Calcipotriene 0.005% Foam and 1% 5-Fluorouracil Cream after Cryotherapy in Treatment of Hyperkeratotic Actinic Keratosis: A Retrospective Study. J. Am. Acad. Dermatol. 2021, 84, 1148–1150. [Google Scholar] [CrossRef]
  285. Dlott, A.; Spencer, S.; Di Pasqua, A. Calcipotriol and 5-Fluorouracil Combination Therapy for the Treatment of Actinic Keratosis in the Clinic: A Review Article. Clin. Drug Investig. 2024, 44, 733–737. [Google Scholar] [CrossRef] [PubMed]
  286. Sonego, B.; Zelin, E.; Bonin, S.; Pozzebon, T.; Bazzacco, G.; Corio, A.; Agozzino, M.; Caro Caposieno, D.R.; Zalaudek, I.; Di Meo, N. Calcipotriol as a Daylight Photodynamic Therapy Enhancer: A Case-Control Study. Dermatol. Rep. 2024, 17, 10077. [Google Scholar] [CrossRef]
  287. Grant, W.B. Roles of Solar UVB and Vitamin D in Reducing Cancer Risk and Increasing Survival. Anticancer Res. 2016, 36, 1357–1370. [Google Scholar] [PubMed]
  288. Feldman, D.; Krishnan, A.V.; Swami, S.; Giovannucci, E.; Feldman, B.J. The role of vitamin D in reducing cancer risk and progression. Nat. Rev. Cancer 2014, 14, 342–357. [Google Scholar] [CrossRef]
  289. Ordonez-Moran, P.; Larriba, M.J.; Pendas-Franco, N.; Aguilera, O.; Gonzalez-Sancho, J.M.; Munoz, A. Vitamin D and cancer: An update of in vitro and in vivo data. Front. Biosci. 2005, 10, 2723–2749. [Google Scholar] [CrossRef]
  290. Slominski, A.T.; Brożyna, A.A.; Skobowiat, C.; Zmijewski, M.A.; Kim, T.K.; Janjetovic, Z.; Oak, A.S.; Jozwicki, W.; Jetten, A.M.; Mason, R.S.; et al. On the role of classical and novel forms of vitamin D in melanoma progression and management. J. Steroid Biochem. Mol. Biol. 2018, 177, 159–170. [Google Scholar] [CrossRef]
  291. Podgorska, E.; Drzal, A.; Matuszak, Z.; Swakon, J.; Slominski, A.; Elas, M.; Urbanska, K. Calcitriol and Calcidiol Can Sensitize Melanoma Cells to Low–LET Proton Beam Irradiation. Int. J. Mol. Sci. 2018, 19, 2236. [Google Scholar] [CrossRef]
  292. Tatiya, A.; Patil, J.; Girase, T.; Patil, M.; Patel, K. An Overview on Management of Psoriasis Using Calcipotriene and Its Amalgamation as Nano Based Drug Delivery System. Mater. Proc. 2023, 14, 38. [Google Scholar] [CrossRef]
  293. Parveen, S.; Sartaj, A.; Iqubal, K.; Kumari, P.; Dang, S.; Aldosari, E.; Ali, J. Topical Co-Delivery of Tacrolimus and Calcipotriol Loaded Nanostructured Lipid Carrier: A Potential and Synergistic Approach in the Management of Psoriasis. J. Dispers. Sci. Technol. 2024, 46, 1301–1314. [Google Scholar] [CrossRef]
  294. Pradhan, M.; Alexander, A.; Singh, M.R.; Singh, D.; Saraf, S.; Saraf, S.; Yadav, K. Statistically Optimized Calcipotriol Fused Nanostructured Lipid Carriers for Effectual Topical Treatment of Psoriasis. J. Drug Deliv. Sci. Technol. 2021, 61, 102168. [Google Scholar] [CrossRef]
  295. Baldwin, H.; Webster, G.; Stein Gold, L.; Callender, V.; Cook-Bolden, F.E.; Guenin, E. 50 Years of Topical Retinoids for Acne: Evolution of Treatment. Am. J. Clin. Dermatol. 2021, 22, 315–327. [Google Scholar] [CrossRef]
  296. Ramchatesingh, B.; Martínez Villarreal, A.; Arcuri, D.; Lagacé, F.; Setah, S.A.; Touma, F.; Al-Badarin, F.; Litvinov, I.V. The Use of Retinoids for the Prevention and Treatment of Skin Cancers: An Updated Review. Int. J. Mol. Sci. 2022, 23, 12622. [Google Scholar] [CrossRef]
  297. Dogra, S.; Yadav, S. Acitretin in Psoriasis: An Evolving Scenario. Int. J. Dermatol. 2014, 53, 525–538. [Google Scholar] [CrossRef] [PubMed]
  298. Carr, D.R.; Trevino, J.J.; Donnelly, H.B. Retinoids for Chemoprophylaxis of Nonmelanoma Skin Cancer. Dermatol. Surg. 2011, 37, 129–145. [Google Scholar] [CrossRef]
  299. Weinstock, M.A.; Bingham, S.F.; Digiovanna, J.J.; Rizzo, A.E.; Marcolivio, K.; Hall, R.; Eilers, D.; Naylor, M.; Kirsner, R.; Kalivas, J.; et al. Tretinoin and the prevention of keratinocyte carcinoma (Basal and squamous cell carcinoma of the skin): A veterans affairs randomized chemoprevention trial. J. Investig. Dermatol. 2012, 132, 1583–1590. [Google Scholar] [CrossRef]
  300. Peck, G.L. Topical tretinoin in actinic keratosis and basal cell carcinoma. J. Am. Acad. Dermatol. 1986, 15, 829–835. [Google Scholar] [CrossRef]
  301. Sami, N.; Feld, S.d.l.; Wolverton, S.E. 46-Topical retinoids. In Comprehensive Dermatologic Drug Therapy, 4th ed.; Elsevier: Amsterdam, The Netherlands, 2021; pp. 528–540.e4. [Google Scholar]
  302. Kang, S.; Goldfarb, M.T.; Weiss, J.S.; Metz, R.D.; Hamilton, T.A.; Voorhees, J.J.; Griffiths, C.E. Assessment of adapalene gel for the treatment of actinic keratoses and lentigines: A randomized trial. J. Am. Acad. Dermatol. 2003, 49, 83–90. [Google Scholar] [CrossRef] [PubMed]
  303. Anaya-Ruiz, M.; Pérez-Santos, M. Drug Repurposing of Adapalene for Melanoma Treatment. Pharm. Pat. Anal. 2022, 11, 9–14. [Google Scholar] [CrossRef]
  304. Rusu, A.; Tanase, C.; Pascu, G.-A.; Todoran, N. Recent Advances Regarding the Therapeutic Potential of Adapalene. Pharmaceuticals 2020, 13, 217. [Google Scholar] [CrossRef]
  305. Wu, C.-S.; Chen, G.-S.; Lin, P.-Y.; Pan, I.-H.; Wang, S.-T.; Lin, S.H.; Yu, H.-S.; Lin, C.-C. Tazarotene Induces Apoptosis in Human Basal Cell Carcinoma via Activation of Caspase-8/t-Bid and the Reactive Oxygen Species-Dependent Mitochondrial Pathway. DNA Cell Biol. 2014, 33, 652–666. [Google Scholar] [CrossRef]
  306. Bianchi, L.; Orlandi, A.; Campione, E.; Angeloni, C.; Costanzo, A.; Spagnoli, L.G.; Chimenti, S. Topical Treatment of Basal Cell Carcinoma with Tazarotene: A Clinicopathological Study on a Large Series of Cases. Br. J. Dermatol. 2004, 151, 148–156. [Google Scholar] [CrossRef]
  307. Tang, J.Y.; Chiou, A.S.; Mackay-Wiggan, J.; Aszterbaum, M.; Chanana, A.M.; Lee, W.; Lindgren, J.; Acosta Raphael, M.; Thompson, B.J.; Bickers, D.R.; et al. Tazarotene: Randomized, Double-Blind, Vehicle-Controlled and Open-Label Concurrent Trials for Basal Cell Carcinoma Prevention and Therapy in Patients with Basal Cell Nevus Syndrome. Cancer Prev. Res. 2014, 7, 292–299. [Google Scholar] [CrossRef] [PubMed]
  308. Cosio, T.; Di Prete, M.; Gaziano, R.; Lanna, C.; Orlandi, A.; Di Francesco, P.; Bianchi, L.; Campione, E. Trifarotene: A Current Review and Perspectives in Dermatology. Biomedicines 2021, 9, 237. [Google Scholar] [CrossRef]
  309. Mahajan, A.; Singh, L.; Singh, G.; Dhawan, R.; Kaur, M.; Malhi, P.; Thakur, K.; Kaur, L. An Evidence-Based Review on Bexarotene. Tumor Discov. 2023, 2, 0436. [Google Scholar] [CrossRef]
  310. Bubna, A. Alitretinoin in Dermatology-An Update. Indian J. Dermatol. 2015, 60, 520. [Google Scholar] [CrossRef] [PubMed]
  311. Sallam, M.A.; Prakash, S.; Krishnan, V.; Todorova, K.; Mandinova, A.; Mitragotri, S. Hyaluronic Acid Conjugates of Vorinostat and Bexarotene for Treatment of Cutaneous Malignancies. Adv. Ther. 2020, 3. [Google Scholar] [CrossRef]
  312. Warda, A.; Staniszewski, L.J.P.; Sabir, Z.L.; Livingston, S.; Sausedo, M.A.; Reshi, S.M.; Ron, E.; Applegate, M.T.; Haddad, D.; Khamisi, M.; et al. Development of Bexarotene Analogs for Treating Cutaneous T-Cell Lymphomas. Cells 2023, 12, 2575. [Google Scholar] [CrossRef] [PubMed]
  313. Shah, K.; Date, A.; Joshi, M.; Patravale, V. Solid Lipid Nanoparticles (SLN) of Tretinoin: Potential in Topical Delivery. Int. J. Pharm. 2007, 345, 163–171. [Google Scholar] [CrossRef]
  314. Dragicevic, N.; Maibach, H. Combined Use of Nanocarriers and Physical Methods for Percutaneous Penetration Enhancement. Adv. Drug Deliv. Rev. 2018, 127, 58–84. [Google Scholar] [CrossRef] [PubMed]
  315. Hafeez, A.; Kazmi, I. Dacarbazine Nanoparticle Topical Delivery System for the Treatment of Melanoma. Sci. Rep. 2017, 7, 16517. [Google Scholar] [CrossRef]
  316. Bhattacharya, S.; Sharma, S. Dacarbazine-Encapsulated Solid Lipid Nanoparticles for Skin Cancer: Physical Characterization, Stability, in-Vivo Activity, Histopathology, and Immunohistochemistry. Front. Oncol. 2023, 13, 1102269. [Google Scholar] [CrossRef] [PubMed]
  317. Cardoso, C.O.; Silva-Carvalho, A.É.; de Mota, I.S.; Lopez, R.F.V.; Cunha-Filho, M.; Saldanha-Araújo, F.; Gratieri, T.; Gelfuso, G.M. Effect of Iontophoresis on Dacarbazine Cutaneous Delivery for Melanoma Topical Treatment. Int. J. Pharm. 2024, 665, 124730. [Google Scholar] [CrossRef]
  318. Vankudre, S.; Shirkoli, N.; Hawaldar, R. Enhanced Delivery of Dacarbazine Using Nanosponge Loaded Hydrogel for Targeted Melanoma Treatment: Formulation, Statistical Optimization and Pre-Clinical Evaluation. J. Pharm. Innov. 2025, 20, 27. [Google Scholar] [CrossRef]
  319. Li, C.; Han, X. Co-Delivery of Dacarbazine and All-Trans Retinoic Acid (ATRA) Using Lipid Nanoformulations for Synergistic Antitumor Efficacy Against Malignant Melanoma. Nanoscale Res. Lett. 2020, 15, 113. [Google Scholar] [CrossRef]
  320. Lei, M.; Wang, J.; Ma, M.; Yu, M.; Tan, F.; Li, N. Dual Drug Encapsulation in a Novel Nano-Vesicular Carrier for the Treatment of Cutaneous Melanoma: Characterization and in Vitro/in Vivo Evaluation. RSC Adv. 2015, 5, 20467–20478. [Google Scholar] [CrossRef]
  321. Yang, X.; Chang, L.; Lin, Q.; Wang, J.; Su, P.; Chen, X.; Yi, Z.; Dong, Y.; Luo, J. Microneedle Pretreatment and Cationic Nanoparticles for Transdermal Delivery of Doxorubicin against Melanoma. J. Drug Deliv. Sci. Technol. 2024, 93, 105417. [Google Scholar] [CrossRef]
  322. Slavkova, M.; Dimitrova, D.; Voycheva, C.; Popova, T.; Spassova, I.; Kovacheva, D.; Yordanov, Y.; Tzankova, V.; Tzankov, B. Composite Hydrogel with Oleic Acid-Grafted Mesoporous Silica Nanoparticles for Enhanced Topical Delivery of Doxorubicin. Gels 2024, 10, 356. [Google Scholar] [CrossRef]
  323. de A Huber, L.; Pereira, T.A.; Ramos, D.N.; Dias de Rezende, L.C.; Emery, F.d.S.; Sobral, L.M.; Leopoldino, A.M.; Lopez, R.F.V. Topical Skin Cancer Therapy Using Doxorubicin-Loaded Cationic Lipid Nanoparticles and Lontophoresis. J. Biomed. Nanotechnol. 2015, 11, 1975–1988. [Google Scholar] [CrossRef]
  324. Guo, M.; Zhou, G.; Liu, Z.; Liu, J.; Tang, J.; Xiao, Y.; Xu, W.; Liu, Y.; Chen, C. Direct Site-Specific Treatment of Skin Cancer Using Doxorubicin-Loaded Nanofibrous Membranes. Chin. Sci. Bull. 2017, 63, 92–100. [Google Scholar] [CrossRef] [PubMed]
  325. Yasmeen, I.; Khan, M.A.; Ali, J.; Baboota, S.; Sartaj, A. Topical Delivery of Mannose Conjugated-Doxorubicin-Berberine Nanostructured Lipid Carrier Gel for Skin Cancer Amelioration: Formulation Optimization, in-Silico, in-Vitro, Ex-Vivo Assessment, and Dermatokinetic Analysis. J. Drug Deliv. Sci. Technol. 2024, 93, 105378. [Google Scholar] [CrossRef]
  326. Ahmed, K.S.; Shan, X.; Mao, J.; Qiu, L.; Chen, J. Derma Roller® Microneedles-Mediated Transdermal Delivery of Doxorubicin and Celecoxib Co-Loaded Liposomes for Enhancing the Anticancer Effect. Mater. Sci. Eng. C 2019, 99, 1448–1458. [Google Scholar] [CrossRef]
  327. Ferrari, G.; Pang, L.Y.; De Moliner, F.; Vendrell, M.; Reardon, R.; Higgins, A.J.; Chopra, S.; Argyle, D. Effective Penetration of a Liposomal Formulation of Bleomycin through Ex-Vivo Skin Explants from Two Different Species. Cancers 2022, 14, 1083. [Google Scholar] [CrossRef]
  328. Hamada, H.; Uesugi, D.; Ishihara, K.; Hosoda, R.; Shimoda, K.; Kuboki, A.; Uchida, N.; Kiriake, Y. Transdermal Delivery of Paclitaxel-Anionic Nanoparticles to Epidermis Layer, Pterostilbene, and Pterostilbene Glycoside, and Their Application for Treatment of Skin Cancer and Wrinkle. Int. J. Curr. Microbiol. Appl. Sci. 2024, 13, 1–7. [Google Scholar] [CrossRef]
  329. Tolentino, S.; Monteiro, M.M.; Saldanha-Araújo, F.; Cunha-Filho, M.; Gratieri, T.; Guerra, E.N.S.; Gelfuso, G.M. Bioadhesive Chitosan Films Loading Curcumin for Safe and Effective Skin Cancer Topical Treatment. Pharmaceutics 2025, 17, 18. [Google Scholar] [CrossRef]
  330. Jose, A.; Labala, S.; Ninave, K.M.; Gade, S.K.; Venuganti, V.V.K. Effective Skin Cancer Treatment by Topical Co-Delivery of Curcumin and STAT3 siRNA Using Cationic Liposomes. AAPS Pharmscitech 2018, 19, 166–175. [Google Scholar] [CrossRef]
  331. Samir, B.; El-Kamel, A.; Zahran, N.; Heikal, L. Resveratrol-Loaded Invasome Gel: A Promising Nanoformulation for Treatment of Skin Cancer. Drug Deliv. Transl. Res. 2024, 14, 3354–3370. [Google Scholar] [CrossRef] [PubMed]
  332. Kalam, M.A.; Ali, R.; Alhowyan, A.A.; Ahmad, A.; Iqbal, M.; Raish, M. Quercetin-Loaded Transliposomal Gel for Effective Management of Skin Cancer: In Vitro and Cell Line Efficacy Studies. J. Drug Deliv. Sci. Technol. 2024, 96, 105659. [Google Scholar] [CrossRef]
  333. Mangla, B.; Kaur, A.; Kumar, P.; Javed, S.; Ahsan, W.; Popli, H. Development of Nanobigel System Combining Apoptosis-Inducing Daidzein and Flaxseed Oil for the Synergistic Treatment of Skin Cancer. J. Drug Deliv. Sci. Technol. 2024, 98, 105843. [Google Scholar] [CrossRef]
  334. Ivanova, N.A. Anti-Melanoma Activity of Green-Produced Nanosilver-Chlorhexidine Complex. Pharmacia 2025, 72, 7. [Google Scholar] [CrossRef]
  335. Himalini, S.; Nallal, U.M.; Razia, M.; Chinnapan, S.; Chandrasekaran, M.; Ranganathan, V.; Gatasheh, M.; Hatamleh, A.; Al-Khattaf, F.; Kanimozhi, S. Antimicrobial, an-Ti-Melanogenesis and Anti-Tyrosinase Potential of Myco-Synthesized Silver Nanoparticles on Human Skin Melanoma SK-MEL-3 Cells. J. King Saud. Univ. Sci. 2022, 34, 101882. [Google Scholar] [CrossRef]
  336. Patil, S.; Khulbe, P.; Nitalikar, M.; Das, K.; BP, M.; Alshehri, S.; Khormi, A.; Almalki, M.; Hussain, S.; Rabbani, S.; et al. Development of Topical Silver Nano Gel Formulation of Bixin: Characterization, and Evaluation of Anticancer Activity. Saudi Pharm. J. 2024, 32, 102125. [Google Scholar] [CrossRef]
Figure 1. Skin pre-cancer and cancer etiology and types. Created in BioRender. Ivanova, N. (2025) https://BioRender.com/a97i800 (accessed on 27 February 2025).
Figure 1. Skin pre-cancer and cancer etiology and types. Created in BioRender. Ivanova, N. (2025) https://BioRender.com/a97i800 (accessed on 27 February 2025).
Pharmaceutics 17 01009 g001
Figure 2. Conventional versus nanoparticulate dermal drug delivery. Created in BioRender. Ivanova, N. (2025) https://BioRender.com/m89a594 (accessed on 5 March 2025).
Figure 2. Conventional versus nanoparticulate dermal drug delivery. Created in BioRender. Ivanova, N. (2025) https://BioRender.com/m89a594 (accessed on 5 March 2025).
Pharmaceutics 17 01009 g002
Table 1. Prodrugs for the topical treatment of skin cancerous or pre-cancerous lesions.
Table 1. Prodrugs for the topical treatment of skin cancerous or pre-cancerous lesions.
DrugProdrug(s)ActivityReference(s)
TemozolomideTemozolomide hexyl esterMelanoma (in vitro and in vivo animal studies)[77]
Proto-Porphyrin IX5-Aminolevulinic acid;
Methyl-5-aminolevulinate;
Butyl-5-aminolevulinate;
Hexyl-5-aminolevulinate
In photodynamic therapy (PDT) of AK and BCC[78,79,80,81,82]
5-Fluorouracil (5-FU)1-Alkylaminocarbonyl 5-FU;
N-Acyloxymethyl derivatives of 5-FU
AK
BCC
SCC
[83,84,85]
Nicotinic acidTetradecyl NicotinateSkin cancer prevention[86]
ImiquimodImiquimod-oleic acidAK
BCC
SCC
Melanoma
[87,88]
Table 2. A comparative table of the various vesicular nano-carriers.
Table 2. A comparative table of the various vesicular nano-carriers.
Type of VesicleCharacteristicsAdvantages Over Liposomes and OverallAdvantages in Dermal Drug DeliveryReferences
LiposomesFlexible uni- or multi-lamellar vesicles composed of phospholipids, water, and cholesterol-Enhanced drug permeation through skin layers[105,106,107,108,109,110]
Ethosomes and transethosomesFlexible nano-sized vesicles composed of phospholipids, water, and ethanol in high concentrations *Better compatibility with chemically unstable APIsEnhanced drug permeation through skin layers;
highly suitable for transdermal drug delivery
[109,110]
TransferosomesHighly deformable nano-sized vesicles composed of phospholipids, water/water—alcoholic mixture, and an edge activator(s) (most often surfactants) **High deformabilityEnhanced drug permeation through skin layers;
highly suitable for transdermal drug delivery
[111,112]
NiosomesNano-sized vesicles composed of nonionic surfactants, water, and cholesterolImproved chemical stability;
improved ability for drug stabilization;
simpler preparation techniques and conditions;
better reproducible technologies;
lower production cost
Moderate enhancement of drug permeation through skin layers;
suitable for topically active dermal formulations
[112,113,114]
PolymersomesNano-sized vesicles composed of amphiphilic block copolymers, surrounding an aqueous coreIncreased loading capacity;
increased chemical stability;
improved ability for drug stabilization;
suitable carriers for biomolecules such as peptides and proteins, incl. enzymes;
adjustable release rate and suitability for controlled drug delivery
Generally, polymersomes are not suitable for dermal drug delivery due to their reduced penetrability; however, a useful application of these vesicles was recently discovered in the topical delivery of photoprotectors to the epidermis for skin cancer prevention [115][115,116,117,118]
InvasomesNano-sized vesicles similar to transferosomes but using terpenes as permeation enhancersHigh deformabilityEnhanced drug permeation through the skin layers[119,120]
PharmacosomesUltra-fine vesicular, micellar, or hexagonal aggregates carrying APIs that are covalently bound to the lipidsHigher entrapment efficiency;
lowered risk of drug leakage
Improved control over drug liberation;
reduced drug toxicity/adverse reactions
[121,122]
Phytosomes (herbosomes)Nano-sized vesicles formed by the hydrogen binding of polyphenols to the hydrophilic choline heads of the liposomal phospholipidsImproved solubility, permeation, stability, and bioavailability of polyphenolsNew perspectives and opportunities for the dermal drug delivery of polyphenols, incl. in skin cancer treatment[123,124]
CubosomesLyotropic nonlamellar liquid crystalline nano-sized vesicles usually comprise glyceryl monooleate, amphiphilic block copolymers, and/or other stabilizing agentsHigher entrapment efficiency;
lowered risk of drug leakage
Bioadhesiveness;
improved localized therapy of skin diseases; reduced drug toxicity/adverse reactions
[125,126]
GlycerosomesNano-sized vesicles composed of phospholipids, water, and glycerol **Higher entrapment efficiency;
longer shelf life
Enhanced drug permeation through the skin layers[127,128]
ChitosomesChitosan-covered liposomesHigher entrapment efficiency;
improved stability
Bioadhesiveness;
Enhanced drug permeation through the skin layers;
Possibly own antibacterial, antifungal, and anti-tumor activity
[129,130,131,132,133]
* transethosomes contain a permeation enhancer in addition. ** other lipids, such as cholesterol, or additional surface-active or polymeric stabilizers, may also be used in the formulation.
Table 3. Nanotechnological advances in the topical delivery of 5-FU.
Table 3. Nanotechnological advances in the topical delivery of 5-FU.
Formulation/Dosage FormNano-Carrier SystemTherapeutic CombinationResult(s)Reference(s)
MicroemulsionMicroemulsionn/aEffectiveness in reducing carcinomatous areas in SCC-induced tissues; sustained drug release; significant skin permeation; minimal toxicity, enhanced patient compliance by reducing treatment duration and adverse reactions[199]
NanoemulsionNanoemulsionn/aEnhanced skin penetration and reduced irritation compared to conventional 5-FU 1% gel[201]
GelChitosan-functionalized nanoemulsionn/aEnhanced topical delivery of 5-FU; promoted retention of 5-FU in skin, and proven non-irritant properties of the nanoemulsion gel[202]
Microneedle-assisted skin deliveryLiposomesn/aEnhanced 5-FU skin penetration and cytotoxicity against SCC; significant increase in 5-FU skin penetration by combining liposomes and microneedle technology[203]
Liposomal suspensionUltradeformable liposomes (transferosomes)n/aEnhanced 5-FU delivery to deeper skin layers[204]
Liposomal suspensionUltradeformable liposomes (transferosomes)ResveratrolEnhanced anti-tumor action of the drug combination of 5-FU and resveratrol against melanoma cells; improved skin permeation by ultradeformable liposomes[205]
Liposomal suspension applied via subcutaneous injection or iontophoresis-assisted topical administrationActive-targeted immunoliposomesCetuximabReduced 5-FU permeation by iontophoresis as compared to injection and potential for reduced systemic side effects; increased 5-FU accumulation in viable epidermis; increased cellular uptake; effectively reduced proliferation of SCC cells[206]
Liposomal suspensionPolymer-coated liposomesn/aSustained release of 5-FU; increased cytotoxicity against epidermoid carcinoma cells as compared to uncoated liposomes[207]
Niosomal suspensionHyaluronic acid-coated niosomesn/aImproved targeting efficiency for 5-FU delivery, controlled drug release, and improved retention in skin[208]
Microwave-assisted delivery of ethosomal suspensionEthosomesn/aEnhanced skin penetration; increased cytotoxicity against human melanoma cells[209]
GelEthosomesn/aEnhanced local bioavailability of 5-FU; reduced skin irritation compared to marketed formulations[210]
GelNLCsCannabidiolImproved tumor remission and survival rate with reduced tumor volume in NMSCs; enhanced skin retention, excellent uptake, and deposition in skin layers[211]
GelNLCsn/aImproved 5-FU permeation and skin retention; reduced skin irritation compared to plain 5-FU gel[212]
GelSLNsn/aEnhanced permeability, sustained release, and cytotoxicity against melanoma and SCC; improved skin retention and permeation, and safety for topical applications[213]
GelSLNsn/aEnhanced 5-FU skin penetration; reduced inflammatory reaction and reduced symptoms of angiogenesis in skin carcinoma-induced mice[214]
SLNs suspensionSLNsn/aImproved cytotoxicity against melanoma cells compared to 5-FU solution; high entrapment efficiency[215]
n/aChitosan nanoparticlesn/aEnhanced encapsulation efficiency for 5-FU delivery; controlled drug release over 24 h; effectiveness against BCC[216]
Nanofiber matChitosan/Polyvinyl alcohol nanofibersn/aEffective treatment of BCC; high encapsulation efficiency and controlled release of 5-FU over 24 h[217]
NanofilmsPolyhydroxyethyl methacrylate/polyhydroxypropyl methacrylate/sodium deoxycholate nanoparticlesIron–platinum nanoparticlesControlled drug release with minimal side effects; anti-proliferative properties against BCC cells[218]
Cream and gelGold nanoparticlesGold nanoparticlesReduced tumor volume in epidermoid carcinoma-bearing mice compared to free 5-FU, confirming enhanced anti-cancer efficacy and improved skin permeability[219]
pH-responsive micellar hydrogelDeoxycholic acid micellesn/aImproved anti-cancer activity against melanoma cells compared to 5-FU alone[220]
Cream base5-FU nanocrystalsn/aImproved efficiency against epidermoid cancer cells as compared to the micro-sized crystals of 5-FU[221]
Nanofibrous scaffoldsNanofibersEtoposideSustained drug release, significant cytotoxicity, and apoptosis against melanoma cells[222]
n/a—not applicable.
Table 4. Nanotechnological advances in the topical delivery of IMQ.
Table 4. Nanotechnological advances in the topical delivery of IMQ.
Formulation/Dosage FormNano-Carrier SystemResult(s)Reference(s)
PatchNLCsImproved drug deposition in deeper skin layers as compared to commercial cream; patches enhanced patient compliance for topical skin cancer treatment[234]
MicroemulsionMicroemulsionLimited ability of the microemulsions to improve the delivery of IMQ over Aldara™ cream[235]
GelMicroemulsion *α-Tocopheryl polyethylene glycol 1000 succinate and oleic acid enhanced IMQ solubility significantly; the micellar formulation improved skin retention and delivery of the drug[236]
MicroemulsionMicroemulsionIMQ 1% microemulsion delivered similar drug quantities to the epidermis as the commercial product with a 5-fold higher dose of 5%; lower risk of systemic absorption compared to the established product[239]
Gel-like microemulsionMicroemulsionEnhanced IMQ delivery to the skin and by a gel-like formulation with a suitable viscosity for topical application viscosity[240]
NanoemulsionNanoemulsionImproved IMQ solubility and drug release; enhanced cytotoxicity against epidermoid carcinoma cell line compared to commercial formulation[241]
Nanosuspension/nanoemulsionLiposomes, nanocrystals, nanoemulsions, lipid nanocapsulesBest results with respect to permeation enhancement were achieved by using an IMQ nanocrystal suspension or nanoemulsion, which demonstrated a three- and five-fold increased drug delivery, respectively, in comparison to the commercially available cream[242]
Liposomal suspensionUltradeformable liposomes (transferosomes)In vitro anti-melanoma activity[243]
Nanostructured formulation (likely of micro- or nanoemulsion type) *pH-responsive micellesIn vitro anti-melanoma activity; improved skin retention in comparison to Imunocare® commercial product; pH-responsive drug release from micelles and possibility for selective drug release in tumor tissues[244]
Ethosomal suspensionEthosomes/transethosomesIncreased permeation rate of IMQ; improved retention and deposition into SC and the deeper epidermal and dermal layers as compared to Aldara™ cream; superior results were obtained with transethosomes (differing from the classic ethosomes by the presence of a permeation enhancer in their composition)[245]
NanosuspensionPolymeric nanoparticles (dextran nanocapsules)Enhanced IMQ transdermal delivery, high encapsulation efficiency, and controlled drug release[246]
Gelβ-Cyclodextrin-based nanospongesEnhanced permeation and retention; sustained release of IMQ; greater inhibitory effect on fibroblast proliferation as compared to the pure drug[247]
Nanosuspension *Chitosan nanocapsulesControlled IMQ release over 24 h; evaluated skin absorption; effective transdermal delivery[248]
* not specified in the original paper but assumed from the given methodology.
Table 5. Novel therapeutic alternatives in the research phase for the topical treatment of skin pre-neoplastic lesions and malignancies.
Table 5. Novel therapeutic alternatives in the research phase for the topical treatment of skin pre-neoplastic lesions and malignancies.
Therapeutic Agent/CombinationMechanism of
Action
Formulation/Dosage FormNanocarrier
System
Result(s)Reference(s)
DacarbazineAlkylating chemotherapeutic agent (usually for parenteral administration)CreamNLCsSuperior anti-proliferative activity of the lipid nanoparticles with dacarbazine in comparison to pure drug against melanoma cell lines[315]
GelSLNsImproved efficacy of the nano-formulation in the treatment of skin tumors induced in rats, as compared to free drug; stable formulation showing minimal side effects and potential in the topical treatment of melanoma[316]
Iontophoresis-assisted applicationn/aIontophoresis appeared as a promising method to enhance the topical delivery of dacarbazine, potentially offering a safer alternative for melanoma treatment by avoiding the adverse effects of systemic administration; the study highlighted the necessity of stabilizing the drug in advance[317]
GelNanosponges (polymeric)Sustained drug release, effective permeation (73%), good biocompatibility, and superior inhibition of melanoma cell proliferation[318]
Dacarbazine + tretioninAn alkylating chemotherapeutic agent combined with RARs/RAXs agonistSLNs nanosuspensionSLNsEffective inhibition of melanoma cell proliferation, induction of remarkable apoptosis, and inhibition of cell cycle progression and cell migration[319]
NanosuspensionTransethosomesSuperior skin permeation and enhanced cytotoxic effects against cutaneous melanoma, indicating potential for effective topical treatment of skin cancer[320]
DoxorubicinAnti-tumor antibiotic, interfering with DNA and RNA synthesis (usually for parenteral administration)Microneedle-assisted deliveryHybrid cationic nanoparticlesTargeted accumulation in subcutaneous melanoma tumors in mice, induction of apoptosis, and suppression of tumor growth[321]
GelOleic acid-grafted mesoporous silica nanoparticlesImproved permeation compared with the hydrogel containing free drug; improved doxorubicin’s cytotoxic effects toward epidermoid carcinoma cells[322]
Iontophoresis-assisted applicationSLNsIncreased epidermal penetration; superior cytotoxic activity against SCC cells compared to pure drug solution; significant improvement in tumor tissue restriction in vivo[323]
Nanofibrous topical implantable delivery deviceCore–shell implantable nanofibrous membranesControlled drug release, sufficient local concentration, allowing dose reduction and minimizing side effects; efficacy against melanoma tumors in vivo (in mice)[324]
Doxorubicin + berberineAnti-tumor antibiotic, interfering with DNA and RNA synthesis, combined with a naturally occurring isoquinoline alkaloid with proven anti-cancer activity and a complex mechanism of actionGelMannose-conjugated NLCsImproved permeation and skin deposition compared to conventional gel; potential for enhanced dual therapeutic approach for skin cancer amelioration based on in silico study[325]
Doxorubicin + celecoxibAnti-tumor antibiotic, interfering with DNA and RNA synthesis, combined with a non-steroidal anti-inflammatory drug, is effective in inhibiting skin cancer developmentMicroneedle-assisted delivery of liposomal gelLiposomesEnhanced in vivo anti-tumor efficacy of the combined liposomal gel against melanoma in comparison to single-drug liposomes; augmented skin penetration in the case of microneedle-assisted delivery[326]
BleomycinAnti-tumor antibiotic, interfering with DNA synthesis (usually for parenteral administration)CreamLiposomesEnhanced skin penetration by the liposomal formulation as compared to the free drug[327]
PaclitaxelChemotherapeutic agents interfere with the normal function of microtubules during cell divisionAnionic bicellesAnionic bicellesEffective penetration in SC; potential for treating skin cancer based on an in vivo study on mouse papillomas[328]
CurcuminA naturally occurring polyphenol with proven anti-proliferative and anti-tumor activity and a complex mechanism of actionBioadhesive film (patch)n/aEnhanced drug penetration into the skin compared to a curcumin solution control; controlled drug release; effective against metastatic melanoma cells after topical application; superior tumor cell inhibition compared to a single dose of radiotherapy[329]
Curcumin + anti-STAT3 siRNAA naturally occurring polyphenol with proven anti-proliferative and anti-tumor activity, combined with gene therapyIontophoresis-assisted applicationLiposomesInhibition of cancer cell growth in mouse melanoma cells, showing a statistically significant difference compared to treatments with either liposomal curcumin or STAT3 siRNA alone; iontophoretic administration demonstrated similar effectiveness in inhibiting tumor progression and STAT3 protein suppression compared with intra-tumoral administration[330]
ResveratrolA naturally occurring polyphenol with proven anti-proliferative and anti-tumor activity and a complex mechanism of actionGelInvasomesHigh skin deposition and potency; high cellular uptake when tested on SCC; proven in vivo effectiveness in Ehrlich-induced mice models[331]
QuercetinA naturally occurring polyphenol with proven anti-proliferative and anti-tumor activity and a complex mechanism of actionGelTransferosomesEnhanced skin permeation; lower cytotoxic concentrations against melanoma cells in comparison to quercetin conventional gel and solution[332]
Diadzein + flaxseed oilA naturally occurring polyphenol with proven anti-proliferative and anti-tumor activity, combined with a rich source of omega-3-polyunsaturated fatty acids sourceNanobigelNanobigelSustained release, improved drug permeation, and induction of apoptosis in epidermoid carcinoma cells[333]
AgNPs + chlorhexidineSilver nanoparticles with intrinsic anti-tumor activity, combined with a broad-spectrum antiseptic agentBioadhesive film (patch)Green tea catechins-synthesized silver nanoparticlesExplicit and selective anti-melanoma activity of the nanosilver complex, fortified in the composition of the adhesive patch[334]
AgNPsSilver nanoparticles with intrinsic anti-tumor activity as a result of a complex mechanism of actionNanosilver suspensionMyco-synthesized silver nanoparticlesDistinct anti-melanogenic activity[335]
AgNPs + bixineSilver nanoparticles with intrinsic anti-tumor activity, combined with a carotenoid with proven anti-proliferative and anti-tumor activity and a complex mechanism of actionGelBixa orellana seed extract-synthesized silver nanoparticlesModerate inhibitory activity against melanoma cancer cell lines; promising anti-cancer activity in vivo[336]
n/a—not applicable.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ivanova, N. Local Chemotherapy of Skin Pre-Neoplastic Lesions and Malignancies from the Perspective of Current Pharmaceutics. Pharmaceutics 2025, 17, 1009. https://doi.org/10.3390/pharmaceutics17081009

AMA Style

Ivanova N. Local Chemotherapy of Skin Pre-Neoplastic Lesions and Malignancies from the Perspective of Current Pharmaceutics. Pharmaceutics. 2025; 17(8):1009. https://doi.org/10.3390/pharmaceutics17081009

Chicago/Turabian Style

Ivanova, Nadezhda. 2025. "Local Chemotherapy of Skin Pre-Neoplastic Lesions and Malignancies from the Perspective of Current Pharmaceutics" Pharmaceutics 17, no. 8: 1009. https://doi.org/10.3390/pharmaceutics17081009

APA Style

Ivanova, N. (2025). Local Chemotherapy of Skin Pre-Neoplastic Lesions and Malignancies from the Perspective of Current Pharmaceutics. Pharmaceutics, 17(8), 1009. https://doi.org/10.3390/pharmaceutics17081009

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