Next Article in Journal
Measuring Bee Effects on Seed Traits of Hybrid Sunflower
Next Article in Special Issue
New Insights into the Latest Advancement in α-Amylase Inhibitors of Plant Origin with Anti-Diabetic Effects
Previous Article in Journal
Seed Germination Ecology of the Medicinal Plant Peganum harmala (Zygophyllaceae)
Previous Article in Special Issue
Gintonin Isolated from Ginseng Inhibits the Epithelial—Mesenchymal Transition Induced by TGF-β in A549 Lung Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

From Plants to Wound Dressing and Transdermal Delivery of Bioactive Compounds

by
Gabriela Olimpia Isopencu
1,
Cristina-Ileana Covaliu-Mierlă
2 and
Iuliana-Mihaela Deleanu
1,*
1
Department of Chemical and Biochemical Engineering, University Politehnica of Bucharest, Polizu Str. 1-7, 011061 Bucharest, Romania
2
Department of Biotechnical Systems, Faculty of Biotechnical Systems Engineering, University Politehnica of Bucharest, 313 Splaiul Independentei, 060042 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Plants 2023, 12(14), 2661; https://doi.org/10.3390/plants12142661
Submission received: 17 May 2023 / Revised: 10 July 2023 / Accepted: 14 July 2023 / Published: 16 July 2023

Abstract

:
Transdermal delivery devices and wound dressing materials are constantly improved and upgraded with the aim of enhancing their beneficial effects, biocompatibility, biodegradability, and cost effectiveness. Therefore, researchers in the field have shown an increasing interest in using natural compounds as constituents for such systems. Plants, as an important source of so-called “natural products” with an enormous variety and structural diversity that still exceeds the capacity of present-day sciences to define or even discover them, have been part of medicine since ancient times. However, their benefits are just at the beginning of being fully exploited in modern dermal and transdermal delivery systems. Thus, plant-based primary compounds, with or without biological activity, contained in gums and mucilages, traditionally used as gelling and texturing agents in the food industry, are now being explored as valuable and cost-effective natural components in the biomedical field. Their biodegradability, biocompatibility, and non-toxicity compensate for local availability and compositional variations. Also, secondary metabolites, classified based on their chemical structure, are being intensively investigated for their wide pharmacological and toxicological effects. Their impact on medicine is highlighted in detail through the most recent reported studies. Innovative isolation and purification techniques, new drug delivery devices and systems, and advanced evaluation procedures are presented.

Graphical Abstract

1. Introduction

The skin, as the body’s largest and most exposed organ, can provide a readily accessible delivery route for therapeutic substances. In fact, the topical application of remedies has been practiced for thousands of years. From Galen, the Father of Pharmacy, who introduced herbal ingredients into the formulation of drugs, the evolution of skin wound dressing/healing (WD) and transdermal delivery (TD) took an interesting path, which continues, and grows nowadays with the help of modern scientific techniques [1].
Most topical drug delivery systems (DDSs) were traditionally designed for the local administration of active pharmaceutical ingredients (APIs), with numerous applications in the field of skin tissue healing [2,3,4]. However, the concept of soft (permeable) drugs that can also affect the tissues/organs beneath the skin originates from Ibn Sina (Avicenna), the great Persian physician and philosopher, born in 980 AD in the village of Afshanah [5,6]. This is now believed to be the oldest conceptualization of transdermal drug delivery (TDD).
Currently, there is a constant search for new materials and new medical devices (wound patches, scaffolds, and transdermal delivery systems) that can provide effective treatment through the unique, controlled, and targeted delivery of APIs. New methods are sought to replace conventional drug delivery routes (gastric, intravenous, intradermal, etc.) with TDs due to their numerous advantages: allowing the direct access of APIs to the bloodstream through the skin and avoiding liver metabolism and the gastrointestinal tract; allowing the homogenous absorption of APIs and increasing pharmaceutical effectiveness; reducing/eliminating drug alterations that can occur in the interaction with patient enzymatic and immune systems; and, equally important, TD is more comfortable, non- or less invasive, and can be easily self-administered [3,7,8,9]. Furthermore, through bionanotechnology, sophisticated devices can be designed to allow drug storage and controlled, on-demand delivery for several days [6,9].
However, of course, there are some disadvantages as well, and major drawbacks that limit, in many cases, the utilization of natural compounds in topical and TDD systems related to both skin and natural compound properties.
As will be briefly detailed further, development strategies involve two major directions: (i) increase skin permeation using chemical penetration enhancers or other more or less invasive methods (electrical, thermal, mechanical, and others), or a combination of methods (microneedles combined with iontophoresis) [10], and (ii) an increase in the bioavailability, stability, and biodistribution of these natural compounds [11].
This review work summarizes the newest outcomes that involve natural compounds of a plant origin as constituents of novel dermal and transdermal drug delivery systems. Major benefits and also important drawbacks associated with their extraction and isolation, as well as stability and bioavailability, are emphasized. This review presents new technologies and modern techniques developed to minimize traditionally known barriers like the complexity of applications, potential adverse effects, and proof of functional changes.
Synthetically, plant constituents, with or without biological activity, are classified based on the metabolic pathways that produce them, and their role in developing TDDS is presented.

2. Natural Compound Transfer Specifics and API Delivery Methods

Dermal and TDD methods, disregarding the principles and formulations, are directly related to skin and drug/API properties, as already mentioned.

2.1. Skin Properties and Drug Transport Mechanism

The skin is a complex three-layer structure comprised of the epidermis, dermis, and hypodermis. It is a two-way protective barrier preventing any external intrusions (microbial, chemical) and, at the same time, the excessive loss of endogenous material. Skin permeability, determined not only by its structure but also by numerous factors like the anatomic site, gender, age, hydration, or external temperature, is mainly associated with Stratum Corneum (SC, the outermost layer), containing a high amount of keratin [7].
Simplistically, TDD means API penetration through the skin at therapeutic rates, but overall, the process, also called percutaneous absorption, is more complicated, described in five main stages. A “vehicle” (transdermal device, cream, or solvent) is often used to incorporate the drug/API. In the first stage, the active molecule is distributed from the vehicle to the hydrolipidic film based on the partition coefficient and molecule’s mobility [12]. The drug is then absorbed into a specific skin layer (penetration step) and partitioned from SC into the aqueous viable epidermis, from where the molecule diffuses to the upper dermis. The permeation from one layer to another further takes place, and finally, the drug enters the bloodstream (the resorption or absorption step) [13,14,15]. Each stage depends on numerous factors and can be differently kinetically characterized, but overall, the transfer is controlled by the molecule’s passive diffusion [12].
In the case of skin wound healing, the mechanisms are more or less the same, as the API is also entering into the bloodstream, but the required pharmaceutical effect is local.
Two major pathways are known to allow some molecule transfer: transepidermal, when a drug permeates into the cells (intracellular) or through the cellular interspaces (transcellular), and transappendageal (via hair follicles or sweat glands) [15,16]. Disregarding the mechanism or the pathway, there are certain physicochemical properties that a potential drug/bioactive substance must meet for a TD, such as [6,17]:
-
Non-toxicity.
-
Low-dose administration requirements.
-
High lipophilicity.
-
Molecular weight below 500 Da.
-
Hydrophilic/hydrophobic balance corresponding to an n-octanol/water partition coefficient (log P) between 1 and 5 (which corresponds to moderate lipophilicity).
-
Acceptable water solubility (0.05 to 1 mg/mL).
-
Melting point below 250 °C.
These requirements, which would naturally allow successful TDD, are also known as the main factors associated with the limited usage and commercial applications of bioactive TDDS up until now.

2.2. Conventional and Modern Topical and TDD Methods

Depending on the technique(s) that allows API to permeate and/or cross the skin, TDD methods are classified as passive or active.
Passive methods, also known as conventional or chemical methods, which include “passive” patch technology, are based on the passive permeation of bioactive molecules. Often these methods are improved via the implementation of penetration/permeation/skin enhancers (usually a chemical or biochemical substance) or supersaturated systems, or other ways, to enhance the mass transfer driving force or to improve skin permeability, allowing for improving dose control. However, TDDs are not fundamentally changed, and the applicability of passive systems remains limited [12,18]. Furthermore, additional drawbacks are associated with permeation enhancer usage [17]:
-
Toxicity and skin irritation.
-
Two-way skin permeabilization (increase in transepidermal water loss).
-
Additional pharmacological effects.
-
Specificity towards a certain drug.
Active (physical) methods, as accurately described by Alkilani et al., “involve the use of external energy to act as a driving force for drug transport across the skin or by physically disrupting the SC” [15]. There is a need for an enhanced TDD, derived naturally together with biotechnological and bioengineering progress, when new (bio)active substances, macromolecules in general, characterized by a low resistance in the gastrointestinal tract, become available for medical and cosmetic therapies [18]. Since the introduction of the first-generation TDDs at the commercial scale in the 1950s, significant advances have been made, and numerous active methods have been tested and/or implemented, as depicted in Figure 1 [16,19]. Among these, the most applied are electrical techniques like electroporation and iontophoresis, mechanical systems like microneedles, or ultrasound-assisted TDDs. Of course, these techniques are not without disadvantages.
High costs, large-scale feasibility, limited loading capacity, stability, or other issues in controlling drug delivery are determining the search for even more advanced techniques. Most recent studies involve integrated chemical and physical approaches (hybrid methods) [20,21].

3. Bioactive Transdermal Patches and Wound Dressing Materials

Transdermal patches (TP) and WD materials are widely used nowadays. These are designed in many forms and configurations. From ancient formulations consisting in ointments bandaged on the skin (Figure 2a1) to state-of-the-art microneedle patches (Figure 2c), the evolution can be considered spectacular [22,23]. One of the first described TDDs (schematically represented in Figure 2a3), and one of the first studies to prove that the limiting step of drug transfer is the diffusion through the skin layer, was introduced by Wurster and Kramer early in 1960, 10 years before the first patent involving continuous drug-rate-controlling delivery was filled [22,24].
By definition, bioactive means having a certain effect on living organisms [25]. A bioactive transdermal patch (BTP) or a WD material can be a structure of a certain complexity to allow the delivery of a bioactive molecule through the skin, directly to the bloodstream. Disregarding the structural complexity, such a patch consists of the following [22,26]:
  • The active component/drug, which could be a natural or a synthetic component with bioactive properties; it can be supplied from a reservoir or from another structure of the patch [27].
  • Backing material, usually made of elastomers, is used to provide patches’ flexibility and protection from the outer environment and water.
  • Drug-releasing membrane/matrix, made of natural/synthetic polymers/elastomers.
  • Adhesive, to keep the patches’ layers together and adhered to the skin; it may also contain the permeation enhancers and/or the drug.
  • The protective liner.
Countless materials/substances are used as either one of these devices’ structural elements, and natural products of a plant origin are included. In fact, medicinal plants have always been an important part of the global medicinal sector. Traditionally, these were used for their curative properties, but nowadays, there are many other ways to use plant materials in the medical field (in general), and as a TP component (in particular). Many examples can be given regarding the use of cell walls’ structural components or plants’ metabolites as constituents in TP development. In our review, we will consider only the most recent ones (with some exceptions), since other reviews cover older investigations.

4. Natural Products from Plants Used in Medical Devices

In a wide meaning, the natural product (NP) is a chemical compound produced by life (including biotic materials, bio-based materials, bodily fluids), while more restrictively, a natural product is an organic compound produced by a living organism [28,29]. Most NPs are highly complex structures, with a large number of sp3 carbon and oxygen atoms and low-nitrogen and halogen atoms, low cLogP values, and molecular rigidity [30]. These characteristics can be regarded, depending on application specificity, as beneficial in the search for new NP-based drugs and formulations.
Simplistically, NPs derived from plants are classified, based on the metabolic pathways that produce them, as primary or secondary metabolites, although there is no clear boundary between them. Their specific metabolic role, structure, and properties allow them to be used as constituents in TP, as will be further detailed.

4.1. Primary Metabolites as Constituents of BTP

Primary metabolites (PM) are organic compounds responsible for photosynthesis, respiration, and plant development (with intrinsic functions). PM include core molecules like amino acids, organic acids, and unsaturated fatty acids, and major macromolecules built based on these: carbohydrates, lipids, and proteins [31]. PM are used in transdermal formulations mainly as constituents of the releasing matrix, and only in a few cases as bioactive components.
In recent years, a great interest has been given to plant mucilages and gums as excellent sources of plant-based primary compounds.

4.1.1. Plant Mucilages

A plant mucilage is a hydrocolloid, containing mainly carbohydrates/monosaccharides, amino acids, glycoproteins, and uranic acid units, in varying concentrations and compositions, depending on the plant source. For a plant, the mucilage represents a vital constituent of the cell, or a part of the cell walls, and can be found in almost all its parts, including the seeds [32,33]. For humans, a mucilage was traditionally used as a gelling and texturing agent in the food industry. In time, it proved to be a valuable and cost-effective natural component source in the biomedical field as well [34]. It is now used as a biodegradable, biocompatible, and non-toxic rate-controlling and matrix-forming agent in TDDS and WD applications, as will be briefly illustrated in the following examples.
In the early stages of DDS, plant mucilages were used as mucoadhesive polymers, due to their chemical characteristics (possessing numerous carboxyl and hydroxyl groups). Conducted studies involving a Mimosa pudica (family: Mimosaceae) mucilage showed positive effects on both the bioadhesion time and drug release percentage [35]. Similar beneficial observations were recently reported for Datura stramonium leaves’ mucilage. Ahad et al. found in a factorial organized study that the mucilage concentration influences the rate and the quantity of Aceclofenac release and also the adhesion time [36].
An interesting valorization of a plant mucilage’s properties (especially charge density here) involves electrospinning, a modern technique applied to obtain nanofibers. Nanofibrillated mats and patches are considered ideal wound healing materials, allowing a sufficient oxygen supply and facile incorporation of the API [37]. A Plantago ovata mucilage was used as a precursor for the preparation of core–shell nanofibers. Biodegradable stimuli-responsive core–shell nanofibers were prepared using the mucilage as a shell solution (outer needle) and PTX-loaded PCPP-CA micelles (Paclitaxel-loaded Cholic acid conjugated to poly (bis (carboxyphenoxy)phosphazene)) as a core solution (inner needle) [38]. This complex nanomaterial, assembled as a patch, was developed to achieve the transdermal controlled delivery of PTX in breast cancer therapy. In vitro studies revealed 180 h of pH-responsive and sustained drug release, and thus, an improved anticancer efficacy compared to free PTX.
In a similar approach, a unique combination of Hibiscus rosa-sinensis leaves’ mucilage (HLM)–PVA–Pectin was electrospun and crosslinked with glutaraldehyde vapor [37]. The obtained “green mat” can be used as a nanofibrillated polymeric scaffold for wound healing. This is just an example of how natural and synthetic polymers can be combined to obtain a hemocompatible and biodegradable product.
Although the use of plants’ mucilage was not that often reported between 2010 and 2020, and their applicability was rather limited, the shift to “natural” sparked a research influx recently. Furthermore, modern techniques of extraction/isolation, characterization, and investigation allowed the identification of secondary metabolites in mucilages, especially when obtained from foliar parts of a plant [39]. Thus, in some cases, a plant mucilage is used solely as a biopolymer with pharmacological activities (antimicrobial, antioxidant), and in other cases, it is the source of the API in a more complex matrix, as can be seen in Table 1.

4.1.2. Plant Gums

Gums are polysaccharides of multiple sugars, with numerous applications in food, pharmaceutical, and medical applications. In most cases, they are naturally occurring, as a result of plants’ protection mechanisms [58]. These are solid in dry environments, and gluey in the presence of humidity. In fact, a major drawback of gums’ usage derives from their uncontrollable rate of hydration and readily-water-soluble characteristic.
Plant-derived gums can be used directly or chemically modified, aiming for customized characteristics of the product. In reported applications, plant gums were used as excipients or matrix-forming materials in mucoadhesive polymeric systems or medical tablets [59,60]. Results indicate that gums could improve mucoadhesive strength or could be a key factor in controlling drug delivery. In a general approach, aiming for applicability not only in pharmaceutical but also in food, medical, or cosmetic industries, versatile hydrogels and biopolymeric nanocarriers were developed based on tragacanth gum or moringa bark gum, as promising controlled DDS [61,62,63,64,65].
Guar gum, which can be obtained from the seeds of Cyamopsis tetragonoloba, was grafted and used in TDD. Thus, membranes for the transdermal controlled release of diclofenac were fabricated using guar gum grafted with acrylic acid, and nanosilica. Slow and sustained drug delivery was achieved due to cage morphology and hydrophobicity in the case of nanocomposites with 1 wt% nanosilica content [66]. In another study, poly(N-isopropylacrylamide)-grafted guar gum, incorporating cellulose nanofibers (CNF), was used for the TD of diltiazem hydrochloride. The obtained biomaterial proved to be a non-irritant to the skin and non-toxic. Similar to nanosilica, 1 wt% CNF determined optimal thermal, mechanical, and barrier properties. A sustained drug release was obtained for several hours (approx. 23% release after 20 h) [67].
In a different approach, cashew gum extracted from Anacardium occidentale was acetylated, and the obtained solution was used to prepare drug-loaded nanoparticles with nanoprecipitation and dialysis techniques [68]. Nanostructures containing diclofenac diethylamine were designed as drug carriers for TD. Both applied methods allowed an efficient drug encapsulation and in vitro 90% TD permeation was obtained in 6 h. Moreover, the TDD could be controlled.
The complex composition of these plant-derived biopolymers allows them, as emphasized by the described applications, to be successfully used in numerous fields as emulsifying, gelling, stabilizing, binding, and filming agents, or even as active functional ingredients [69]. As promising as it may be, the usage of mucilages and gums has some important limitations to be considered and overcome, aiming for industrial applicability. Disregarding the plant source/plant parts, their composition, which is always very complex, may differ due to climate conditions, regional characteristics, agricultural and agronomical variations, local availability (in some cases), and preparation techniques. The storage conditions must be very carefully addressed, to minimize or eliminate the risk of contamination and microbial spoilage, thermal degradation, and natural biodegradability.

4.1.3. Plant Cell Wall Components

A plant cell wall can be defined as the greatest renewable resource available. It is a complex, heterogenous structure, comprised of more than 90% polysaccharides, specifically assembled, depending on plant species and tissue types [70,71]. Major cell wall structural components that are mainly used as excipients in medical devices are cellulose, hemicellulose, and pectin.
Cellulose, the main component of cell walls of any plant, is chemically defined as unbranched β-(1,4)-linked glucan chains. It presents in a complex inhomogeneous assembly due to multiscale linear fibrils formed during numerous and various biosynthesis processes [71]. As a water insoluble carbohydrate, it was extensively used in many industrial fields. In modern pharmaceuticals, cellulose (as fibrils, microcrystals, or nanocrystals) and cellulose derivates obtained with chemical routes are often used as excipients for the development of controlled DDS [3,72,73,74].
Cellulose nanofibers (CNf), obtained with mechanical methods or pretreatments, are used to increase matrix hydrophilicity, biocompatibility, stability, and mechanical strength [75,76]. Moreover, CNf high porosity allows the facile preparation of 3D network hydrogels that can embody drugs or other active nanoparticles [77]. Similarly, to increase mechanical properties of WD materials, microcrystalline cellulose (MCC) can be used. For instance, Ding et al. prepared a novel fiber-type dressing material composed of alginate, polyvinyl alcohol, MCC, and Euphorbia humifusa Willd extract by microfluidic spinning. The obtained medical dressing proved to have good mechanical properties due to MCC and an antibacterial effect due to the plant extract [78].
Carboxymethyl cellulose (CMC) is the first and maybe the most used among cellulose derivates, due to its characteristics (cost effectiveness, biocompatibility, stability, stimuli responsiveness, and water solubility). Numerous applications and materials were developed based on CMC [79]. Some of the most recent can be named (2023 publication year):
-
Self-healing biodegradable 3D network hydrogel, based on boronic-acid-grafted CMC, used as a drug release vehicle or scaffold for tissue regeneration or bleeding control [80].
-
Antibacterial photo-inspired WD, based on CMC as the “network backbone” hydrogel, polyvinyl alcohol (PVA) with iodophor (KI-I2) as the antimicrobial hydrogel, and a fluorescent material (carbon quantum dots) as the doping agent. This functional material releases desired doses of an antibacterial agent (I2), and provides real-time information on the wound healing process, based on hydrogel color responsiveness to wound pH changes in UV and visible light [81].
-
Multilayer microneedles, based on PVA and CMC for the transdermal delivery of nicotinamide mononucleotide (antiaging agent). CMC properties, as a high water solubility and hydrophilicity, determine a higher drug release [82].
-
Dissolvable patch, based on sodium alginate and CMC, loaded with cephalexin monohydrate as an antimicrobial agent for skin and chronic wound infections. A sustained drug release is obtained for selected concentrations [83].
-
Chromogenic aerogel-like composite, based on CMC and PVA with an anthocyanin extract immobilized into the polymeric matrix, as a color indicator for wound pH and antimicrobial agent at the same time [84].
Other WD materials were developed based on cellulose acetate [85,86,87,88], hydroxyethyl cellulose [89], ethyl cellulose [90], hydroxypropyl methylcellulose [91], or cellulose micro/nanocrystals [78,92] (references are of this year). An interesting approach in developing vegetable patches was reported by Buonvino et al., who used the Lupinus albus L. hull, a vegetal waste, as raw material for the development of a novel active bioplastic, to be used as WD material or a cellular scaffold, benefiting from both cellulosic and phytochemical content of the hulls [93].
Hemicellulosic polysaccharides (β-glycan backbones) are used in developing WD, due to their biocompatibility and biodegradability, but not as much as cellulose [94]. Recently, Li et al. used hemicellulose as a reinforcing component in developing a flexible hydrogel with exceptional characteristics, to be used as self-powered TDD with motion sensing capacity [95].
Pectins are cell wall polysaccharides with α-(1,4)-linked galacturonic acids in their backbone [71]. Their distinctive properties among biopolymers, such as gelling and mucoadhesivity, combined with the resistance to amylases and proteases, encouraged pectin-based nanomaterial development for numerous medical applications, including WD [96]. Pectins are used in a native form or chemically modified. Thus, hydrogels composed of quaternized chitosan and pectin from a citrus (family Rutaceae) peel, containing propolis as an antimicrobial factor, are proposed as WD materials. Both pectin and propolis concentration influenced propolis release profiles, film mechanical properties, and stability [97]. Based on the expertise regarding extensive research, Raghavendra Naveen et al. obtained the extended release of neomycin sulphate from a novel carrier transdermal system—a polymer-layered silicate nanocomposite, developed using polyethyleneglycol, montmorillonite, and thiolated pectin. For an optimized composition, almost 90% of neomycin is released in 18 h. The nanocomposite exhibits the desired properties for a successful TDD (water vapor permeability, swelling, non-cytotoxicity) [98].

4.2. Secondary Metabolites as Constituents of BTP

Based on PM, plants secrete some biologically active chemicals, which are not involved in plants’ development and growth, called secondary metabolites (SM). The function of these SM is mainly to prevent diseases in living organisms, although the function of many of them still remains unknown. Secondary plant metabolites can be classified into four major classes [31,99]: alkaloids, phenolic compounds, sulfur-containing compounds, and terpenoids.
In general, TP containing plant extracts are composed of active principles that activate blood circulation, together with strong irritant substances. It is thus essential to control the concentration of these compounds in the TD system, so that the optimal clinical dose is delivered.

4.2.1. Alkaloids

Alkaloids are a group of nitrogenous heterocyclic bases of a low molecular weight, obtained from different parts of a plant. They are residual alkaline products of plant metabolism, mainly produced in defense mechanisms. Alkaloids present a large spectrum of pharmacological effects and are used as medications or as recreational drugs. Depending on the pharmaceutical effects, they can be classified as local anesthetics and stimulants (cocaine), psychedelics (psilocin), neuro-stimulants (caffeine, nicotine), analgesics (morphine), antibacterial (berberine, kokusaginine, nkolbisine), anticancer drugs (vinblastine, vincristine), antihypertensive agents (reserpine), cholinomimerics (galantamine), spasmolysis agents (atropine), vasodilators (vincamine), antiarrhythmia (quinidine), antiasthma therapeutics (ephedrine), and antimalarials (quinine) [100].
However, not all of these alkaloids can be used as API in TD. Those that are already integrated into medical instruments (TP), or those studied to be used for transdermal treatments, are listed as follows.
Scopolamine is a tropane alkaloid produced by the Solanaceae (nightshade) plant family, such as henbane (Hyoscyamus niger), devil’s trumpet (Datura stramonium), angel’s trumpet (Brugmansia versicolor), deadly nightshade (Atropa belladonna), mandrake (Mandragora officinarum), and corkwood (Duboisia myoporoides) [100]. It was the first approved active substance impregnated in a TP for commercial use (by FDA in 1979). It is used to ameliorate motion sickness and nausea [101,102] or for its anticholinergic action [103]. The principle of passive transport through skin layers was maintained unchanged until the present, as well as the following manufacturing principles [104]:
(a)
An adhesive layer that sticks to the skin and contains scopolamine in a polymeric gel, which provides an initial priming dose.
(b)
An intermediate microporous polypropylene rate-controlling membrane.
(c)
The scopolamine reservoir that sustains a zero-order input of a drug to the skin surface.
(d)
A backing of impermeable aluminized polyester film.
Opiates (morphine, codeine) are isoquinoline alkaloids extracted or refined from natural plant matter—poppy sap and fibers (Papaver somniferum). Since morphine is among the first analgesic substances used, it is also the first tested for TD, to reduce the metabolic effects of ingestion, or the discomfort of intravenous injection. Westerling, 1994 proposed a rudimentary transdermal morphine delivery system. The system involves bypassing the SC by actually removing the SC using a vacuum. This leads to the formation of a lesion on the skin, over which a diffusion chamber containing gauze impregnated with morphine is applied. The diffusion yield is 75% in the first 11 h after application [105]. Although the diffusion rate is quite low and the clinical effect is also low, TP with morphine have been developed. Thus, a TP made of polyethylene sponge foam as the retaining agent was designed to be applied directly to the skin. Morphine hydrochloride is delivered without any invasive permeation enhancer or a rate-limiting membrane. An adhesive layer to attach the patch to the clean, non-hairy dermal surface is also involved [106,107].
Caffein is a purin-alkaloid found in more than 60 species of plants across the globe. Caffeine comes from the seeds of coffee beans (Coffea arabica and Coffea robusta), cacao beans (Theobroma cacao), and Kola nuts (Cola acuminata, Cola nitida); the leaves and buds of tea; the leaves of Yerba mate (Ilex paraguariensis); or the bark of Yoco (Paullinia yoco) [100]. This alkaloid acts as a vasodilator involved in numerous nervous system disorders: migraine, epilepsy, traumatic brain injury, ischemia, anxiety, alcoholism, Alzheimer’s disease, and brain cancer [108,109]. Because of a low solubility in the lipid layer of the SC, caffeine can be a TD using different penetration enhancers [110,111], or physical penetration methods to cross the SC of the skin (mechanical, ultrasounds, ionophoresis, microneedles) [108,111,112,113,114,115].
Nicotine is a potent parasympathomimetic alkaloid mostly found in Solanaceae. It is found in a high concentration (approximately 0.6 to 3.0% of the dry weight) in Nicotiana spp. (tobacco), and is present in the range of 2 to 7 μg/kg in various edible plants of the Solanaceae family such as tomatoes (genera Lycopersicon), potatoes and eggplants (genera Solanum), and peppers (genera Capsicum) [100]. There are a multitude of commercial TP containing nicotine, used in particular to reduce smoking addiction or migraines. The active substance is incorporated in the form of a drug reservoir, or impregnated in a matrix [26,116].
Berberine hydrochloride (BH), or berberine, is an isoquinoline alkaloid extracted from several plants of the genus Berberis such as Berberis aquifolium, Berberis vulgaris, Berberis aristata (Berberidaceae), and other genera including Coptis chinensis, Hydrastis canadensis, Xanthorhiza simplicissima (Ranunculaceae), Phellodendron amurense (Rutaceae), Tinospora cordifolia (Menispermaceae), Argemone mexicana, and Eschscholzia californica (Papaveraceae) [100]. It has a strong antimicrobial ability, broad antibacterial spectrum, and wide applicability prospect in the treatment of bacterial and fungal infections and other skin diseases [117]. The reduced solubility in both aqueous and lipidic environments makes berberine require other substances or methods to facilitate TD [118]. But, the importance of this alkaloid in the treatment of skin diseases, including psoriasis, has motivated a multitude of studies regarding appropriate trans epidermal transport methods and supports (nanocarrier, the impregnation of nanobiocellulose of a microbial origin, etc.) [119,120].
Galantamine hydrobromide (GH) is an alkaloid that proves effective therapeutic management in memory impairment diseases, especially Alzheimer’s disease, but recent studies used this alkaloid as transdermal treatment for rheumatoid arthritis [121,122]. Galantamine is found in various plant sources, mainly from the genera Amaryllis, Lycoris, Hippeastrum, Ungernia, Leucojum, Zephyranthes, Narcissus, Galanthus, Hymenocallis, and Haemanthus, and is a naturally occurring alkaloid of the family Amaryllidaceae [123]. GH was originally isolated from plants, but now is chemically synthesized.
Ephedrine is an alkaloid with a phenethylamine skeleton, obtained from the plant Ephedra sinica and other members of the genus Ephedra, from where the name of the substance derives [124]. Ephedrine causes cardiovascular effects similar to those of epinephrine: an increase of blood pressure, heart rate, and contractility. Like pseudoephedrine, it is a bronchodilator, but with a considerably higher effect. Ephedrine can reduce motion sickness, but it is mainly used to reduce the sedative effects of other motion sickness drugs combined in TP [125,126,127].

4.2.2. Phenolic Compounds (PC)

Phenolic compounds (PC) are SM widely distributed in the plant kingdom, found in various higher organs of different plants, such as medicinal plants, spices, vegetables, and cereals, with about 8000 different phenolic structures [128,129,130,131,132,133,134].
The most important role of these substances is that of plant adaptation in stressful conditions, such as damages, infections, or exposure to UV radiation, but they also may have secondary roles such as involving color, aroma, taste, etc. [128,132,135]. The presence of PC in the diet and in medical treatments is recognized in the prevention of many human diseases. Research carried out to date demonstrates the benefits of PC to the human body, such as antioxidant, antimicrobial, vasoactive, and anti-inflammatory effects, their ability to interact with enzymes and cellular receptors, etc. [136,137,138,139,140].
From a structural point of view, PC are substances that contain at least one benzene ring (C6) and one or more hydroxyl groups [128,135]. The metabolic pathways through which plants produce these SM are (i) the shikimate/chorismate or succinyl–benzoate pathway, which produces phenylpropanoid derivatives (C6-C3); (ii) the acetate/malonate or polyketide pathway, which produces side-chain elongated phenylpropanoids, including the large group of flavonoids (C6-C3-C6) and some quinones; and (iii) the acetate/mevalonate pathway, which produces aromatic terpenoids, mostly monoterpenes, through dehydrogenation reactions [128,135,141,142].
There are different criteria to classify PC, by the number of carbon atoms in the molecule, by the number of phenol units, or even according to the importance in the human diet [141,142]. Most classifications consider only the number of phenolic units in the structure of PC; however, some consider both the structure of PC and their pharmacological importance. In this review, the classification adopted by Nurzynska-Wierdak, 2023 is considered the most appropriate to describe the importance of PC in the dermal and TD of various human ailments. Thus, PC are divided into five subgroups [142]:
-
Phenolic acids (hydroxybenzoic and hydroxycinnamic acids).
-
Flavonoids (flavonols and flavan-3-ols, flavones, flavanones, isoflavones, flavanones and anthocyanidins).
-
Coumarins.
-
Lignans.
-
Stilbenes.
The bioavailability of PC is not very good; therefore, their inclusion in controlled drug release systems is a current research topic. Problems such as a poor solubility, stability, and permeability are to be overcome. The bioavailability of PC depends on the subclass of phenolic compounds and their physicochemical properties, such as the degree of polymerization or molecular properties, polarity, and interactions with different components of the formulation in which they are introduced [138,139,143].

Non-Flavonoid PC

Phenolic acids (PA) (hydroxybenzoic acids—HBA and hydroxycinnamic acids—HCA)
HBAs have a C6-C1 main structure derived directly from benzoic acid and include p-HBAs such as proto-catechuic, vanillic, gallic, and syringic acids, while HCAs are aromatic compounds with a side chain of three carbon atoms (C6-C3), and they are represented by coumaric, caffeic, ferulic, and synaptic acids. PA are found in vegetables, and specific HBAs are abundantly found in oilseeds, cereals, coffee beans, cowpeas, blackcurrants, raspberries, skins and seeds of pumpkins, and blackberries, while HCAs are found in coffee beans, cherries, grains, peaches, spinach, citrus fruits, plums, tomatoes, potatoes, and almonds [142,144,145].
PA have both antioxidant and antimicrobial action, but there are not many studies on their individual action in TDD, specifically in TP. Most often, they are investigated together with other phenolic compounds, as natural plant extracts, in the transdermal treatment of wound healing as ion gels, nanoemulgels (anti-inflammatory treatments, skin disorders, diabetic gangrene), or microneedles for better TD [110,146,147,148,149,150,151,152].
Curcuminoids are bright yellow pigments, representing a group of lipophilic diketones extracted from the rhizomes of Curcuma longa, a plant from the Zingiberaceae family, used for medicinal purposes for decades mostly in the Asian region. Curcumin, the main used curcuminoid, with antitumoral, antioxidant, antiarthritic, anti-amyloid, anti-ischemic, and anti-inflammatory properties, is poorly absorbed through skin barriers. Because of its hydrophobicity, low bioavailability, and chemical instability, different formulations are studied to improve its therapeutic efficacity including as nanoemulsions, nanohydrogels, nanoparticles in smart film and nanoparticles in hydrogels, nanostructured lipid carriers, lipid-based self-nanoemulsifying systems, ethosomes, and microneedles [90,153,154,155,156,157,158,159,160,161].
Lignans are dimeric structures formed by a β-β′ covalent bond between two phenylpropanoid units. Neo-lignans are a subclass of lignans in which the covalent bond is replaced by any other type of connection, including oxygen etheric linking. All natural compounds that co-occur with lignans or neo-lignans, and possess C15, C16, or C17 core structures, are named nor-lignans [162]. Lignans show a large array of biological properties, including antioxidant, antifungal, antibacterial, and anti-inflammatory properties [163,164,165,166,167]. The most common biological source of lignans is flax (Linum usitatissimum) seeds [162,168]. However, these bioactive compounds are very lipophilic and their potential use in pharmacological studies is limited, because of a very low solubility in aqueous media [169,170]. The TD of lignans can be facilitated by sophorolipid-based transferosomal hydrogel or microemulgel in cosmetic application, or lignan-loaded solid lipid nanoparticles for topical application [170,171,172].
Stilbenes belong to polyphenol group compounds and consist of two phenyl groups connected by ethene. They are found in fruit crops (especially grapes, peanuts, berries) and medicinal plants, including Polygonum multiflorum, Polygonum cuspidatum (Polygonaceae), Hopea chinensis (Dipterocarpaceae), Gnetum parvifolium (Gnetaceae), Caragana sinica (Leguminosae), Morus alba (Moraceae), etc. The most frequently used stilbene derivate compounds are pterostilbene (PTS) and resveratrol (RSV). Exploited now in medicine, stilbenes possess a variety of biological activities, including antitumoral, anti-inflammatory, antioxidant, antibacterial, and antiviral activities [11,165,173,174,175,176]. Several systems have been studied for the TD of stilbenes, such as polyvinyl pyrrolidone-based dissolving microneedle patches, or different types of nanocarriers: anionic phospholipids, polymeric nanoparticles, lipid base nanoparticles, cyclodextrin complexation, electrospun nanofibers, nanocrystals, liposomes, dendrimers, and niosomes [11,174,177,178,179,180].

Flavonoid PC

Flavonoid PC are a class of substances composed of more than 6000 low-molecular-weight phenolic compounds with a skeleton of flavan. The main subgroups are flavones, flavonols, flavonones, flavononols, flavan-3-ols, anthocyanines, isoflavones, and chalcones. Flavonoids are substances with strong antioxidant, anti-inflammatory, and antiplatelet activities and are found in a very wide range of vegetables and fruits [181,182,183]. Among the many delivery systems developed so far, lipid-based nanoparticles, including liposomes and lipid nanoparticles, as well as polymer-based nanoparticles, are the most commonly used for the TD of flavonoids, because of their lipophilic nature and poor water solubility, which lead to a limited bioavailability.
The studies carried out on the use of flavonoids in TP focus in particular on the following subclasses:
-
Flavanols represented by a catechin group, containing epicatechin (EG) and specifically epigallocatechin gallate (EGCG), have attracted much attention due to their wide spectrum of biological properties, including antioxidant, photoprotective, antiviral, and antibacterial, as well as anticarcinogenic and neuroprotective, properties. EGCG is used as TP in the treatment of atopic dermatitis in the form of loaded microneedles, or with Ag nanoparticles in cotton patches for gangrene in diabetics [184,185]; EG is a TD using microneedles [114].
-
Flavanones, especially naringenin and hesperidin, are inserted into TP with an anti-inflammatory purpose. They are used for atopic dermatitis, WD nanoemulsions or nanogels on different polymeric supports, or lipidic nanocarriers [186,187,188].
-
Flavones represented by apigenin, which is a hydrophobic flavone, have been used in several formulations so far for TD, including liposomes, nanocrystal gel formulations, and self-micro-emulsifying delivery systems for anti-inflammatory and breast cancer treatment [189,190]. Luteolin is another promising flavonoid with antiarthritic activity. Topical formulations to treat psoriasis, or to keep breast cancer under control, were developed [191,192]. Due to its lipolytic character, the transport system is of a lipidic nanocarrier or niosome type [192,193,194].
-
Flavonols—quercetin is one of the most intensively studied and common flavonols found in nature; because this flavonoid shows a poor permeability in normal or excised human skin, it has been incorporated into various delivery systems, including nanoemulsions, nanocapsules, lipid nanoparticles, and microemulsions, to increase skin solubility and permeability [139,195]. TP with quercetin, incorporated in nanocomposite materials using microneedles, are used for the treatment of inflammation or androgenic alopecia [134,196]. Kaempferol is another well-known flavonol with antioxidant, anti-inflammatory, anticancer, and antiallergic properties, and it is used in the same TD systems compared to other flavonoids, mainly using lipid nanocarriers [197].
-
Isoflavones—represented mainly by genistein and daidzein, found in soybeans (Glycine max) and mung beans (Vigna radiata). Both are a TD with lipid nanocarriers [198,199].
For the TD of flavonoids, Zhang and Michniak-Kohn (2020) introduced the term “flavosomes”, the new deformable liposomes, which, in addition to the flavonoid compound, with the lipolytic structure, can be associated with an active synthetic API, to potentiate the anti-inflammatory action.
Additionally, a special class of flavonoids, intensively studied for TD in inflammation, cardiovascular diseases, or skin conditions, such as acne, is the licorice flavonoids, extracted from the root and rhizomes of the Glycyrrhiza glabra (Leguminosae family). These contain the same classes as the typical flavonoids, and their most important representatives include liquiritin, isoliquiritin, liquiritigenin, isoliquiritigenin, glabridin, licochalcone A, licoflavone A, licochalcone B, retrochalcone, and formononetin [194,200]. The TD of this type of flavonoids is achieved through microemulsions or hydrogels [194,201].

4.2.3. Sulfur-Containing Compounds

Sulfur-containing compounds can also be isolated from plants, and used for their diverse biological activities, such as anti-inflammatory, antioxidant, and anticancer activities [202,203,204,205,206,207,208].
Organosulfur natural products (ONP) refer to types of organic natural products that contain sulfur elements, such as thiols, thioesters, sulfoxides, etc., which play an important role in the pharmaceutical industry [209,210]. Although studies focus mainly on glucosinolates and S-alk(en)yl cysteine sulfoxides, there are other sulfur-containing secondary metabolites synthesized from plants that are of research interest (e.g., H2S, SO2, antimicrobial peptides, etc.) [211]. Glucosinolates are non-reactive sulfur-containing phytoanticipins found in plants of the family Brassicaceae (Brassica oleracea species, e.g., broccoli, cabbage, cauliflower, etc.), and S-alk(en)yl cysteine sulfoxides (ASCOs) are predominantly found in the Allium family of vegetables (i.e., Allium sativum, Allium cepa, Allium porrum), and their thiosulfinated derivatives are behind many of the flavor compounds of these vegetables [212]. Natural organic sulfur compounds are used in TDD combined with other pharmacological substances in lipid base nanocarriers for the treatment of wounds and psoriasis, as well as for antibacterial or antitumoral activity [213,214].

4.2.4. Terpenoids

Terpenoids—the largest class of plant SM represents about 60% of all known natural products. Over 40,000 different identified structures of terpenoids, or isoprenoids, are found in plants, and they are responsible for the scents, flavors, and colors [215]. In a TD system, essential oils (EO), included in this group, represent the most efficient systems for SC penetration and transdermal transport facilitation, being called “natural enhancers” [216,217,218,219,220].
This class of SM basically consists of five carbon isoprene units, assembled to each other (many isoprene units) in thousands of ways. Terpenes are simple hydrocarbons, while terpenoids are a modified class of terpenes, with isopentenyl pyrophosphate (IPP) as a structural unit, with different functional groups and an oxidized methyl group moved or removed at various positions [221].
Based on the number of isoprene units, terpenoids can be classified into hemiterpenoids (one isoprene unit—C5), and their occurrence is rare and they have no special biological significance; monoterpenoids (two isoprene units—C10); sesquiterpenoids (three isoprene units—C15); diterpenoids (four isoprene units—C20); sesterterpenoids (five isoprene units—C25); triterpenoids (six isoprene units—C30); tetraterpenoids (eight isoprene units—C40); and other terpenoid (5nC) isoprene unit condensation compounds [215,222]. As follows, the terpenoids used in WD and TDS are presented in brief.
-
Hemiterpenes are the simplest terpenes, and the number of known compounds is lower than 100. Most hemiterpenes are in the form of oils and insoluble in water, with some glycosylated exceptions that are water soluble. The best-known hemiterpene is isoprene, which is the basic unit of all terpenes. Other known examples are tiglic, angelic acids, and isovaleric acid. These were used as enhancers for SC permeability in TP developed based on poly 3-hydroxybutyric acid-co-3-hydroxy valeric acid (PHBV) nanofibers, to deliver active substances (e.g., curcumin) for WD and to ensure an antimicrobial effect [223].
-
Monoterpenes are widely distributed in secretory tissues such as oil glands or resin chambers and ducts of higher plants. Monoterpenes are the major constituents of volatile plant oils. A number of monoterpenes are oxygenated and many exhibit biological and perfuming activities. Some are often used as medicinal agents or ingredients in cosmetic products. Monoterpenes frequently used as SC penetration agents are anethol, borneol, and campho (used in commercial TP for pain release); carvacol, carvone, and 1.8–1.4 cineol (used for TDDS in psychiatry); cymene, eugenol, fenchone, geraniol, limonene, linalool, and menthol (used as an enhancer and also for pain release); menthone, a-pinen oxide, pulegone, rose oxide, safranal, terpinene-4-ol, a-terpineol, tetra-hydrogeneraniol, and tymol (oral patches); and verbenone (perfumery and cosmetic use) [215,224,225,226,227,228,229,230,231,232,233,234,235,236,237,238,239,240,241,242].
-
Sesquiterpenes are the class of SM consisting of three isoprene units (C15) and are found in linear, cyclic, bicyclic, and tricyclic forms. Since sesquiterpenes have a high allergenic potential on the skin, their use in TP is limited [243,244,245].
-
Diterpenoids belong to a versatile class of chemical constituents found in different natural sources having four isoprene units. They can be classified as linear, bicyclic, tricyclic, tetracyclic, pentacyclic, or macrocyclic diterpenes, depending on their skeletal core. In nature, they are commonly found in a polyoxygenated form, with keto and hydroxyl groups, often esterified by small-sized aliphatic or aromatic acids [244]. This class of compounds show significant biological activities including anti-inflammatory, antimicrobial, anticancer, antifungal, cardiovascular, etc. [215]. Among tricyclic diterpenoids, abietic acid was used for wound healing and tanshinone IIA—with numerous therapeutic uses—was also included in TD systems as nanoparticles or microneedles [246,247,248].
-
Triterpenoids are a major class of SM and contain compounds with a carbon skeleton based on six isoprene units, derived biosynthetically from the acyclic C30 hydrocarbon, squalene. They have relatively complex cyclic structures, most being either alcohols, aldehydes, or carboxylic acids [215]. Sterols are triterpenes that are based on the cyclopentane perhydrophenanthrene ring system. Plant sterols called “phytosterols”, e.g., sitosterol, stigmasterol, and campesterol, are widespread in higher plants. Triterpenes have many active sites for glycosylation, which converts them into another class of compounds, namely, saponins (triterpene glycoside). Examples of triterpenoids of medical interest are squalene used in scaffold emulgel for wound healing, ursolic acid used in niosomal gel systems for arthritis treatment and skin diseases, oleanolic acid used in nanocarriers in TD, and sitosterol used in alopecia treatment with microneedle enhancers, or in breast cancer as nanoparticles [249,250,251,252,253,254,255].
-
Tetraterpenoids consist of eight isoprene units and 25 carbon atoms. The most common tetraterpenoids are carotenoids, which are natural fat soluble pigments [244]. Carotenoids are extremely hydrophobic molecules, with little or no solubility in water, and a strong tendency to aggregate in aqueous solutions. Since carotenoids are unstable at high temperatures, in the presence of light and oxygen, their incorporation into micro- and nanostructures is necessary, to increase their stability and preserve their bioactivity [256,257]. Examples of used carotenoids are as follows: lycopene loaded in a lipid nanocarrier in TDD systems; β-carotene in TDS as self-nanoemulsion or as lipid base nanocarriers, or as hydrogel with plant extracts; lutein with a high antioxidant effect is delivered in TDS using nanostructured lipid carriers, solid lipid nanoparticles, a nanosphere, a liposome, nanoemulsions, and polymeric nanoparticles for eye diseases, for anti-inflammatory effects, obesity, etc.; and crocin in a nanocellulose membrane of microbial cellulose-based TP [258,259,260,261,262,263,264].
-
Polyterpenoids are polymeric isoprenoid hydrocarbons, which consist of more than eight isoprene units. This class of compounds include rubber and gutta percha, used as polymeric matrices for TDDS [16,265].
Table 2, presented as follows, highlights the most recent medical applications (only 2023 publication year) of SM derived from plants, in topical or TD treatments, for various ailments or wound treatment.

5. Conclusions

Currently, medical devices for transdermal delivery and wound dressing materials are of a wide diversity, especially regarding the way of the transport and delivery of the active pharmaceutical ingredients to the skin and through the Stratum Corneum. Plants’ primary and secondary metabolites are solely used or combined for a favorable medical diagnosis, both at the level of the skin and endogenously targeted at certain types of tissues.
This review shows how plants were broken down into active principles or nonactive substances, and according to the different types of these compounds’ classes, how they were used for the treatment of various ailments. In ancient times, a plant was used as such (fresh or dried), or infused and impregnated on various textile supports, thus contributing with its entire arsenal of primary and secondary metabolites, as well as related microbiota, to the treatment of various diseases. Presently, technological possibilities allow the advanced extraction and purification of active compounds from plants, which contribute to a more efficient assembly of medical devices, better transdermal transport yields, and well-established orientation towards the target.
As new separation and preparation technologies can now offer a high purity and nanosized drugs, it is certain that new delivery vectors, and consequently effective treatment systems, should and can be developed in the near future. In a constantly increasing market of skin patches and transdermal drug delivery devices, interdisciplinary efforts are further required to develop strategies for the easier and safer administration of natural bioactive compounds.

Author Contributions

Equal contribution of all authors for conceptualization, investigation, resources, and writing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Raju, N.S.; Krishnaswami, V.; Vijayaraghavalu, S.; Kandasamy, R. Transdermal and Bioactive Nanocarriers. In Nanocosmetics: Fundamentals, Applications and Toxicity; Elsevier Inc.: Amsterdam, The Netherlands, 2020; pp. 17–33. ISBN 9780128222867. [Google Scholar]
  2. Cao, X.; Sun, L.; Luo, Z.; Lin, X.; Zhao, Y. Aquaculture Derived Hybrid Skin Patches for Wound Healing. Eng. Regen. 2023, 4, 28–35. [Google Scholar] [CrossRef]
  3. Raghav, N.; Sharma, M.R.; Kennedy, J.F. Nanocellulose: A Mini-Review on Types and Use in Drug Delivery Systems. Carbohydr. Polym. Technol. Appl. 2021, 2, 100031. [Google Scholar] [CrossRef]
  4. Sousa, M.P.; Neto, A.I.; Correia, T.R.; Miguel, S.P.; Matsusaki, M.; Correia, I.J.; Mano, J.F. Bioinspired Multilayer Membranes as Potential Adhesive Patches for Skin Wound Healing. Biomater. Sci. 2018, 6, 1962–1975. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Amr, S.S.; Tbakhi, A. Ibn Sina (Avicenna): The Prince Of Physicians. Ann. Saudi Med. 2007, 27, 134–135. [Google Scholar] [CrossRef] [PubMed]
  6. Szunerits, S.; Boukherroub, R. Heat: A Highly Efficient Skin Enhancer for Transdermal Drug Delivery. Front. Bioeng. Biotechnol. 2018, 6, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Peña-Juárez, M.C.; Guadarrama-Escobar, O.R.; Escobar-Chávez, J.J. Transdermal Delivery Systems for Biomolecules. J. Pharm. Innov. 2022, 17, 319–332. [Google Scholar] [CrossRef] [PubMed]
  8. Kuznetsova, D.A.; Vasilieva, E.A.; Kuznetsov, D.M.; Lenina, O.A.; Filippov, S.K.; Petrov, K.A.; Zakharova, L.Y.; Sinyashin, O.G. Enhancement of the Transdermal Delivery of Nonsteroidal Anti-Inflammatory Drugs Using Liposomes Containing Cationic Surfactants. ACS Omega 2022, 7, 25741–25750. [Google Scholar] [CrossRef] [PubMed]
  9. Verma, P.; Maheshwari, N.; Bairagee, D. Physiochemical Characterization and in Vitro Evaluation of Formulated Herbal Bioactive Loaded Transdermal Patches. Pharmacogn. Res. 2022, 15, 64–70. [Google Scholar] [CrossRef]
  10. Mohammed, Y.; Holmes, A.; Kwok, P.C.L.; Kumeria, T.; Namjoshi, S.; Imran, M.; Matteucci, L.; Ali, M.; Tai, W.; Benson, H.A.E.; et al. Advances and Future Perspectives in Epithelial Drug Delivery. Adv. Drug Deliv. Rev. 2022, 186, 114293. [Google Scholar] [CrossRef]
  11. Chavda, V.P.; Patel, A.B.; Mistry, K.J.; Suthar, S.F.; Wu, Z.X.; Chen, Z.S.; Hou, K. Nano-Drug Delivery Systems Entrapping Natural Bioactive Compounds for Cancer: Recent Progress and Future Challenges. Front. Oncol. 2022, 12, 867655. [Google Scholar] [CrossRef]
  12. Richard, C.; Cassel, S.; Blanzat, M. Vesicular Systems for Dermal and Transdermal Drug Delivery. RSC Adv. 2020, 11, 442–451. [Google Scholar] [CrossRef] [PubMed]
  13. Ita, K. Transcutaneous Drug Administration. In Transdermal Drug Delivery; Academic Press: Cambridge, MA, USA, 2020; pp. 1–7. [Google Scholar] [CrossRef]
  14. Sabbagh, F.; Kim, B.S. Recent Advances in Polymeric Transdermal Drug Delivery Systems. J. Control. Release 2022, 341, 132–146. [Google Scholar] [CrossRef]
  15. Alkilani, A.Z.; McCrudden, M.T.C.; 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] [Green Version]
  16. 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]
  17. Kováčik, A.; Kopečná, M.; Vávrová, K. Permeation Enhancers in Transdermal Drug Delivery: Benefits and Limitations. Expert Opin. Drug Deliv. 2020, 17, 145–155. [Google Scholar] [CrossRef] [PubMed]
  18. Brown, M.B.; Martin, G.P.; Jones, S.A.; Akomeah, F.K. Dermal and Transdermal Drug Delivery Systems: Current and Future Prospects. Drug Deliv. J. Deliv. Target. Ther. Agents 2006, 13, 175–187. [Google Scholar] [CrossRef] [Green Version]
  19. Prausnitz, M.R.; Langer, R. Transdermal Drug Delivery. Nat. Biotechnol. 2008, 26, 1261–1268. [Google Scholar] [CrossRef] [PubMed]
  20. Alkilani, A.Z.; Nasereddin, J.; Hamed, R.; Nimrawi, S.; Hussein, G.; Abo-Zour, H.; Donnelly, R.F. Beneath the Skin: A Review of Current Trends and Future Prospects of Transdermal Drug Delivery Systems. Pharmaceutics 2022, 14, 1152. [Google Scholar] [CrossRef] [PubMed]
  21. Tapfumaneyi, P.; Imran, M.; Mohammed, Y.; Roberts, M.S. Recent Advances and Future Prospective of Topical and Transdermal Delivery Systems. Front. Drug Deliv. 2022, 2, 957732. [Google Scholar] [CrossRef]
  22. Pastore, M.N.; Kalia, Y.N.; Horstmann, M.; Roberts, M.S. Transdermal Patches: History, Development and Pharmacology. Br. J. Pharmacol. 2015, 172, 2179–2209. [Google Scholar] [CrossRef] [Green Version]
  23. Jiang, X.; Xia, W.; Pan, J.; Yang, W.; Zhang, S.; Li, C.; Zan, T.; Lai, Y.; Xu, Z.; Yu, H. Engineered Microneedle Systems for Topical Cancer Therapy. Appl. Mater. Today 2023, 31, 101774. [Google Scholar] [CrossRef]
  24. Wurster, D.E.; Kramer, S.F. Investigation of Some Factors Influencing Percutaneous Absorption. J. Pharm. Sci. 1961, 50, 288–293. [Google Scholar] [CrossRef] [PubMed]
  25. Bioactive Definition & Meaning-Merriam-Webster. Available online: https://www.merriam-webster.com/dictionary/bioactive (accessed on 12 April 2023).
  26. Bird, D.; Ravindra, N.M. Transdermal Drug Delivery and Patches—An Overview. Med. Devices Sens. 2020, 3, e10069. [Google Scholar] [CrossRef]
  27. Wang, C.; Bai, J.; Tian, P.; Xie, R.; Duan, Z.; Lv, Q.; Tao, Y. The Application Status of Nanoscale Cellulose-Based Hydrogels in Tissue Engineering and Regenerative Biomedicine. Front. Bioeng. Biotechnol. 2021, 9, 732513. [Google Scholar] [CrossRef] [PubMed]
  28. Abozenadah, H.; Bishop, A.; Bittner, S.; Lopez, O.; Wiley, C.; Flatt, P.M. CH105: Chapter 6-A Brief History of Natural Products and Organic Chemistry-Chemistry. In Consumer Chemistry: How Organic Chemistry Impacts Our Lives; Western Oregon University: Monmouth, OR, USA, 2017. [Google Scholar]
  29. Cheng, Y.C.; Li, T.S.; Su, H.L.; Lee, P.C.; Wang, H.M.D. Transdermal Delivery Systems of Natural Products Applied to Skin Therapy and Care. Molecules 2020, 25, 5051. [Google Scholar] [CrossRef] [PubMed]
  30. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Orhan, I.E.; Banach, M.; Rollinger, J.M.; Barreca, D.; Weckwerth, W.; Bauer, R.; Bayer, E.A.; et al. Natural Products in Drug Discovery: Advances and Opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  31. Adetunji, C.O.; Palai, S.; Ekwuabu, C.P.; Egbuna, C.; Adetunji, J.B.; Ehis-Eriakha, C.B.; Kesh, S.S.; Mtewa, A.G. General Principle of Primary and Secondary Plant Metabolites: Biogenesis, Metabolism, and Extraction. In Preparation of Phytopharmaceuticals for the Management of Disorders: The Development of Nutraceuticals and Traditional Medicine; Elsevier Inc.: Amsterdam, The Netherlands, 2020; pp. 3–23. ISBN 9780128202845. [Google Scholar]
  32. Dybka-Stępień, K.; Otlewska, A.; Góźdź, P.; Piotrowska, M. The Renaissance of Plant Mucilage in Health Promotion and Industrial Applications: A Review. Nutrients 2021, 13, 3354. [Google Scholar] [CrossRef]
  33. Goksen, G.; Demir, D.; Dhama, K.; Kumar, M.; Shao, P.; Xie, F.; Echegaray, N.; Lorenzo, J.M. Mucilage Polysaccharide as a Plant Secretion: Potential Trends in Food and Biomedical Applications. Int. J. Biol. Macromol. 2023, 230, 123146. [Google Scholar] [CrossRef]
  34. Filho, J.G.D.O.; Lira, M.M.; de Sousa, T.L.; Campos, S.B.; Lemes, A.C.; Egea, M.B. Plant-Based Mucilage with Healing and Anti-Inflammatory Actions for Topical Application: A Review. Food Hydrocoll. Health 2021, 1, 100012. [Google Scholar] [CrossRef]
  35. Ahuja, M.; Kumar, S.; Yadav, M. Evaluation of Mimosa Seed Mucilage as Bucoadhesive Polymer. Yakugaku Zasshi 2010, 130, 937–944. [Google Scholar] [CrossRef] [Green Version]
  36. Ahad, H.A.; Chinthaginjala, H.; Sai Priyanka, M.; Raghav, D.R.; Gowthami, M.; Naga Jyothi, V. Datura Stramonium Leaves Mucilage Aided Buccoadhesive Films of Aceclofenac Using 32 Factorial Design with Design-Expert Software. Indian J. Pharm. Educ. Res. 2021, 55, s396–s404. [Google Scholar] [CrossRef]
  37. Sen, S.; Bal, T.; Rajora, A.D. Green Nanofiber Mat from HLM–PVA–Pectin (Hibiscus Leaves Mucilage–Polyvinyl Alcohol–Pectin) Polymeric Blend Using Electrospinning Technique as a Novel Material in Wound-Healing Process. Appl. Nanosci. 2022, 12, 237–250. [Google Scholar] [CrossRef] [PubMed]
  38. Mehnath, S.; Chitra, K.; Karthikeyan, K.; Jeyaraj, M. Localized Delivery of Active Targeting Micelles from Nanofibers Patch for Effective Breast Cancer Therapy. Int. J. Pharm. 2020, 584, 119412. [Google Scholar] [CrossRef] [PubMed]
  39. Saju, F.; Sivaraman, C.M. Scope of Herbal Mucilage in Pharmaceutical Formulations. A Review. Herba Pol. 2021, 67, 46–57. [Google Scholar] [CrossRef]
  40. Ahad, H.A.; Kumar, C.S.; Ravindra, B.V.; Sasidhar, C.G.S.; Ramakrishna, G.; Venkatnath, L.; Gangadhar, P.; Navya, K. Characterization and Permeation Studies of Diltiazem Hydrochloride-Ficus Reticuleta Fruit Mucilage Transdermal Patches. Int. J. Pharm. Sci. Rev. Res. 2010, 1, 32–37. [Google Scholar]
  41. Rangari, N.T.; Kalyankar, T.M.; Puranik, P.K.; Chaudhari, S.R. Permeation Studies of Pioglitazone HCL from Ficus Carica Fruit Mucilage Matrix Transdermal Patches. Int. J. Pharm. Sci. Res. 2012, 3, 3927–3931. [Google Scholar] [CrossRef]
  42. Ahad, H.A.; Ishaq, B.M.; Shaik, M.; Bandagisa, F. Designing and Characterizing of Tramadol Hydrochloride Transdermal Patches Prepared with Ficus Carica Fruit Mucilage and Povidone. Pak. J. Pharm. Sci. 2016, 29, 945–951. [Google Scholar]
  43. Siddique, A.; Ratnamala, K.V. Formulation and Evaluation of Naproxen Sodium Transdermal Patch Using Natural Polymers. Hum. J. Res. Artic. Dec. 2017, 11, 160–176. [Google Scholar]
  44. Marthadu, S.; Medindla, J.; Sreelekha, B.; Nishkala, B. Aceclofenac Penetration Studies from Transdermal Patch Prepared with Ficus Benghalensis Fruit Mucilage as Matrix Forming Polymer. Int. J. Pharma Sci. Res. 2020, 11, 8–11. [Google Scholar]
  45. Baral, P.; Bhattarai, A.; Shahi, L.; Shrestha, R.; Adhikari, S.; Giri, A. Formulation and Characterization of Diclofenac Potassium Transdermal Patch-Es Prepared with Ficus Auriculata Fruit Mucilage and Hydroxypropyl Methyl Cellulose K4M. J. Pharm. Drug Dev. 2020, 7, 1–11. [Google Scholar]
  46. Hosseini, M.S.; Nabid, M.R. Synthesis of Chemically Cross-Linked Hydrogel Films Based on Basil Seed (Ocimum Basilicum L.) Mucilage for Wound Dressing Drug Delivery Applications. Int. J. Biol. Macromol. 2020, 163, 336–347. [Google Scholar] [CrossRef] [PubMed]
  47. Tantiwatcharothai, S.; Prachayawarakorn, J. Property Improvement of Antibacterial Wound Dressing from Basil Seed (O. Basilicum L.) Mucilage-ZnO Nanocomposite by Borax Crosslinking. Carbohydr. Polym. 2020, 227, 115360. [Google Scholar] [CrossRef] [PubMed]
  48. Sadat Hosseini, M.; Kamali, B.; Nabid, M.R. Multilayered Mucoadhesive Hydrogel Films Based on Ocimum Basilicum Seed Mucilage/Thiolated Alginate/Dopamine-Modified Hyaluronic Acid and PDA Coating for Sublingual Administration of Nystatin. Int. J. Biol. Macromol. 2022, 203, 93–104. [Google Scholar] [CrossRef]
  49. Izadyari Aghmiuni, A.; Heidari Keshel, S.; Sefat, F.; Akbarzadeh Khiyavi, A. Quince Seed Mucilage-Based Scaffold as a Smart Biological Substrate to Mimic Mechanobiological Behavior of Skin and Promote Fibroblasts Proliferation and h-ASCs Differentiation into Keratinocytes. Int. J. Biol. Macromol. 2020, 142, 668–679. [Google Scholar] [CrossRef] [PubMed]
  50. Oran, D.; Unal, S.; Gunduz, O. Evaluation of Bacterial Cellulose/Quince Seed Mucilage Composite Scaffold for Wound Dressing. Emergent Mater. 2022, 5, 315–321. [Google Scholar] [CrossRef]
  51. Nilani, P.; Pranavi, A.; Duraisamy, B.; Damodaran, P.; Subhashini, V.; Elango, K. Assessment of Wound Healing Potential of Dermal Patches. Int. J. Pharm. Pharmacol. 2021, 10, 001–006. [Google Scholar]
  52. Ramos, I.F.d.S.; Magalhães, L.M.; O Pessoa, C.D.; Ferreira, P.M.P.; Rizzo, M.D.S.; Osajima, J.A.; Silva-Filho, E.C.; Nunes, C.; Raposo, F.; Coimbra, M.A.; et al. New Properties of Chia Seed Mucilage (Salvia Hispanica L.) and Potential Application in Cosmetic and Pharmaceutical Products. Ind. Crop. Prod. 2021, 171, 113981. [Google Scholar] [CrossRef]
  53. Massey, S.; Iqbal, F.; Rehman, A.U.; Iqbal, M.S.; Iram, F. Preparation, Characterization and Biological Evaluation of Silver Nanoparticles and Drug Loaded Composites for Wound Dressings Formed from Lallemantia Royleana Seeds’ Mucilage. J. Biomater. Sci. Polym. Ed. 2022, 33, 481–498. [Google Scholar] [CrossRef]
  54. Hassan, I.; Gani, A.; Ahmad, M.; Banday, J. Extraction of Polysaccharide from Althea Rosea and Its Physicochemical, Anti-Diabetic, Anti-Hypertensive and Antioxidant Properties. Sci. Rep. 2022, 12, 17116. [Google Scholar] [CrossRef]
  55. Pereira, R.; Carvalho, A.; Vaz, D.C.; Gil, M.H.; Mendes, A.; Bártolo, P. Development of Novel Alginate Based Hydrogel Films for Wound Healing Applications. Int. J. Biol. Macromol. 2013, 52, 221–230. [Google Scholar] [CrossRef]
  56. Nair, R.S.; Ling, T.N.; Abdul Shukkoor, M.S.; Manickam, B. Matrix Type Transdermal Patches of Captopril: Ex Vivo Permeation Studies through Excised Rat Skin. J. Pharm. Res. 2013, 6, 774–779. [Google Scholar] [CrossRef]
  57. Chelu, M.; Musuc, A.M.; Aricov, L.; Ozon, E.A.; Iosageanu, A.; Stefan, L.M.; Prelipcean, A.M.; Popa, M.; Moreno, J.C. Antibacterial Aloe Vera Based Biocompatible Hydrogel for Use in Dermatological Applications. Int. J. Mol. Sci. 2023, 24, 3893. [Google Scholar] [CrossRef] [PubMed]
  58. Rana, V.; Rai, P.; Tiwary, A.K.; Singh, R.S.; Kennedy, J.F.; Knill, C.J. Modified Gums: Approaches and Applications in Drug Delivery. Carbohydr. Polym. 2011, 83, 1031–1047. [Google Scholar] [CrossRef]
  59. George, B.; Suchithra, T.V. Plant-Derived Bioadhesives for Wound Dressing and Drug Delivery System. Fitoterapia 2019, 137, 104241. [Google Scholar] [CrossRef]
  60. Ray, P.; Chatterjee, S.; Saha, P. Screening of Polysaccharides from Fruit Pulp of Ziziphus Mauritiana L. and Artocarpus Heterophyllus L. as Natural Mucoadhesives. Futur. J. Pharm. Sci. 2021, 7, 29. [Google Scholar] [CrossRef]
  61. Sadat Hosseini, M.; Hemmati, K.; Ghaemy, M. Synthesis of Nanohydrogels Based on Tragacanth Gum Biopolymer and Investigation of Swelling and Drug Delivery. Int. J. Biol. Macromol. 2016, 82, 806–815. [Google Scholar] [CrossRef]
  62. Pathania, D.; Verma, C.; Negi, P.; Tyagi, I.; Asif, M.; Kumar, N.S.; Al-Ghurabi, E.H.; Agarwal, S.; Gupta, V.K. Novel Nanohydrogel Based on Itaconic Acid Grafted Tragacanth Gum for Controlled Release of Ampicillin. Carbohydr. Polym. 2018, 196, 262–271. [Google Scholar] [CrossRef]
  63. Sheorain, J.; Mehra, M.; Thakur, R.; Grewal, S.; Kumari, S. In Vitro Anti-Inflammatory and Antioxidant Potential of Thymol Loaded Bipolymeric (Tragacanth Gum/Chitosan) Nanocarrier. Int. J. Biol. Macromol. 2019, 125, 1069–1074. [Google Scholar] [CrossRef]
  64. Gupta, V.K.; Sood, S.; Agarwal, S.; Saini, A.K.; Pathania, D. Antioxidant Activity and Controlled Drug Delivery Potential of Tragacanth Gum-Cl-Poly (Lactic Acid-Co-Itaconic Acid) Hydrogel. Int. J. Biol. Macromol. 2018, 107, 2534–2543. [Google Scholar] [CrossRef]
  65. Roy, D.; Bal, T.; Swain, S. Fabrication and Evaluation of PH-Sensitive Biocompatible Microwave Irradiated Moringa Barkgum-Carrageenan (MOG-CRG-IPN) Interpenetrating Isotropic Polymeric Network for Controlled Delivery of Pharmaceuticals. Sustain. Chem. Pharm. 2020, 18, 100325. [Google Scholar] [CrossRef]
  66. Giri, A.; Bhunia, T.; Mishra, S.R.; Goswami, L.; Panda, A.B.; Pal, S.; Bandyopadhyay, A. Acrylic Acid Grafted Guargum-Nanosilica Membranes for Transdermal Diclofenac Delivery. Carbohydr. Polym. 2013, 91, 492–501. [Google Scholar] [CrossRef]
  67. Dutta, K.; Das, B.; Orasugh, J.T.; Mondal, D.; Adhikari, A.; Rana, D.; Banerjee, R.; Mishra, R.; Kar, S.; Chattopadhyay, D. Bio-Derived Cellulose Nanofibril Reinforced Poly(N-Isopropylacrylamide)-g-Guar Gum Nanocomposite: An Avant-Garde Biomaterial as a Transdermal Membrane. Polymer 2018, 135, 85–102. [Google Scholar] [CrossRef]
  68. Dias, S.F.L.; Nogueira, S.S.; De França Dourado, F.; Guimarães, M.A.; De Oliveira Pitombeira, N.A.; Gobbo, G.G.; Primo, F.L.; De Paula, R.C.M.; Feitosa, J.P.A.; Tedesco, A.C.; et al. Acetylated Cashew Gum-Based Nanoparticles for Transdermal Delivery of Diclofenac Diethyl Amine. Carbohydr. Polym. 2016, 143, 254–261. [Google Scholar] [CrossRef] [PubMed]
  69. Tosif, M.M.; Najda, A.; Bains, A.; Kaushik, R.; Dhull, S.B.; Chawla, P.; Walasek-Janusz, M. A Comprehensive Review on Plant-Derived Mucilage: Characterization, Functional Properties, Applications, and Its Utilization for Nanocarrier Fabrication. Polymers 2021, 13, 1066. [Google Scholar] [CrossRef] [PubMed]
  70. Houston, K.; Tucker, M.R.; Chowdhury, J.; Shirley, N.; Little, A. The Plant Cell Wall: A Complex and Dynamic Structure as Revealed by the Responses of Genes under Stress Conditions. Front. Plant Sci. 2016, 7, 984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Zhang, B.; Gao, Y.; Zhang, L.; Zhou, Y. The Plant Cell Wall: Biosynthesis, Construction, and Functions. J. Integr. Plant Biol. 2021, 63, 251–272. [Google Scholar] [CrossRef]
  72. Mohd Zuki, S.A.; Abd Rahman, N.; Abu Bakar, N.F. Nanocrystal Cellulose as Drug Excipient in Transdermal Patch for Wound Healing: An Overview. IOP Conf. Ser. Mater. Sci. Eng. 2018, 334, 012046. [Google Scholar] [CrossRef]
  73. Latif, M.S.; Azad, A.K.; Nawaz, A.; Rashid, S.A.; Rahman, M.H.; Al Omar, S.Y.; Bungau, S.G.; Aleya, L.; Abdel-Daim, M.M. Ethyl Cellulose and Hydroxypropyl Methyl Cellulose Blended Methotrexate-Loaded Transdermal Patches: In Vitro and Ex Vivo. Polymers 2021, 13, 3455. [Google Scholar] [CrossRef]
  74. Manna, S.; Dhanalakshmi, D.; Bhowmik, M.; Jana, S.; Jana, S. Cellulose Derivative-Based Bioadhesive Blend Patch for Transdermal Drug Delivery. Front. Mater. 2022, 9, 835507. [Google Scholar] [CrossRef]
  75. Koochaki, A.; Shahgholi, M.; Sajadi, S.M.; Babadi, E.; Inc, M. Investigation of the Mechanical Stability of Polyethylene Glycol Hydrogel Reinforced with Cellulose Nanofibrils for Wound Healing: Molecular Dynamics Simulation. Eng. Anal. Bound. Elem. 2023, 151, 1–7. [Google Scholar] [CrossRef]
  76. Razack, S.A.; Lee, Y.; Shin, H.; Duraiarasan, S.; Chun, B.S.; Kang, H.W. Cellulose Nanofibrils Reinforced Chitosan-Gelatin Based Hydrogel Loaded with Nanoemulsion of Oregano Essential Oil for Diabetic Wound Healing Assisted by Low Level Laser Therapy. Int. J. Biol. Macromol. 2023, 226, 220–239. [Google Scholar] [CrossRef] [PubMed]
  77. Dong, D.; Chen, R.; Jia, J.; Zhao, C.; Chen, Z.; Lu, Q.; Sun, Y.; Huang, W.; Wang, C.; Li, Y.; et al. Tailoring and Application of a Multi-Responsive Cellulose Nanofibre-Based 3D Nanonetwork Wound Dressing. Carbohydr. Polym. 2023, 305, 120542. [Google Scholar] [CrossRef]
  78. Ding, M.; Wang, X.; Man, J.; Li, J.; Qiu, Y.; Zhang, Y.; Ji, M.; Li, J. Antibacterial and Hemostatic Polyvinyl Alcohol/Microcrystalline Cellulose Reinforced Sodium Alginate Breathable Dressing Containing Euphorbia Humifusa Extract Based on Microfluidic Spinning Technology. Int. J. Biol. Macromol. 2023, 239, 124167. [Google Scholar] [CrossRef]
  79. Pourmadadi, M.; Rahmani, E.; Shamsabadipour, A.; Samadi, A.; Esmaeili, J.; Arshad, R.; Rahdar, A.; Tavangarian, F.; Pandey, S. Novel Carboxymethyl Cellulose Based Nanocomposite: A Promising Biomaterial for Biomedical Applications. Process Biochem. 2023, 130, 211–226. [Google Scholar] [CrossRef]
  80. Zhang, K.; Wu, D.; Chang, L.; Duan, W.; Wang, Y.; Li, W.; Qin, J. Cellulose Based Self-Healing Hydrogel through Boronic Ester Connections for Wound Healing and Antitumor Applications. Int. J. Biol. Macromol. 2023, 230, 123294. [Google Scholar] [CrossRef] [PubMed]
  81. Yang, Y.; Zhang, C.; Gong, M.; Zhan, Y.; Yu, Z.; Shen, C.; Zhang, Y.; Yu, L.; Chen, Z. Integrated Photo-Inspired Antibacterial Polyvinyl Alcohol/Carboxymethyl Cellulose Hydrogel Dressings for PH Real-Time Monitoring and Accelerated Wound Healing. Int. J. Biol. Macromol. 2023, 238, 124123. [Google Scholar] [CrossRef] [PubMed]
  82. Sabbagh, F.; Kim, B.-S. Ex Vivo Transdermal Delivery of Nicotinamide Mononucleotide Using Polyvinyl Alcohol Microneedles. Polymers 2023, 15, 2031. [Google Scholar] [CrossRef]
  83. Feketshane, Z.; Adeyemi, S.A.; Ubanako, P.; Ndinteh, D.T.; Ray, S.S.; Choonara, Y.E.; Aderibigbe, B.A. Dissolvable Sodium Alginate-Based Antibacterial Wound Dressing Patches: Design, Characterization, and in Vitro Biological Studies. Int. J. Biol. Macromol. 2023, 232, 123460. [Google Scholar] [CrossRef]
  84. Alsahag, M.; Alisaac, A.; Al-Hazmi, G.A.A.; Pashameah, R.A.; Attar, R.M.S.; Saad, F.A.; El-Metwaly, N.M. Preparation of Carboxymethyl Cellulose/Polyvinyl Alcohol Wound Dressing Composite Immobilized with Anthocyanin Extract for Colorimetric Monitoring of Wound Healing and Prevention of Wound Infection. Int. J. Biol. Macromol. 2023, 224, 233–242. [Google Scholar] [CrossRef]
  85. Aldossary, H.A.; Khalaf, M.M.; Gouda, M.; Elmushyakhi, A.; Abou Taleb, M.F.; Abd El-Lateef, H.M. Wound Dressing Candidate Materials Based on Casted Films of Cellulose Acetate Modified with Zirconium Oxide (ZrO2), and Gallium Oxide (Ga2O3). Mater. Today Commun. 2023, 34, 105299. [Google Scholar] [CrossRef]
  86. Sathe, T.; Bodas, D. Development and Characterization of a Polydimethylsiloxane-Cellulose Acetate Hybrid Membrane for Application in Organ-on-a-Chip. Mater. Sci. Eng. B Solid-State Mater. Adv. Technol. 2023, 291, 116366. [Google Scholar] [CrossRef]
  87. Kumar, G.; Khan, F.G.; Abro, M.I.; Aftab, U.; Jatoi, A.W. Development of Cellulose Acetate/CuO/AgNP Nanofibers Based Effective Antimicrobial Wound Dressing. Compos. Commun. 2023, 39, 101550. [Google Scholar] [CrossRef]
  88. Graça, M.F.P.; Melo, B.L.; Lima-Sousa, R.; Ferreira, P.; Moreira, A.F.; Correia, I.J. Reduced Graphene Oxide-Enriched Chitosan Hydrogel/Cellulose Acetate-Based Nanofibers Application in Mild Hyperthermia and Skin Regeneration. Int. J. Biol. Macromol. 2023, 229, 224–235. [Google Scholar] [CrossRef] [PubMed]
  89. Le, K.T.; Nguyen, C.T.; Lac, T.D.; Nguyen, L.G.T.; Tran, T.L.; Tran-Van, H. Facilely Preparing Carboxymethyl Chitosan/Hydroxyethyl Cellulose Hydrogel Films for Protective and Sustained Release of Fibroblast Growth Factor 2 to Accelerate Dermal Tissue Repair. J. Drug Deliv. Sci. Technol. 2023, 82, 104318. [Google Scholar] [CrossRef]
  90. Nawaz, A.; Farid, A.; Safdar, M.; Latif, M.S.; Ghazanfar, S.; Akhtar, N.; Al Jaouni, S.K.; Selim, S.; Khan, M.W. Formulation Development and Ex-Vivo Permeability of Curcumin Hydrogels under the Influence of Natural Chemical Enhancers. Gels 2022, 8, 384. [Google Scholar] [CrossRef]
  91. Natori, N.; Shibano, Y.; Hiroki, A.; Taguchi, M.; Miyajima, A.; Yoshizawa, K.; Kawano, Y.; Hanawa, T. Preparation and Evaluation of Hydrogel Film Containing Tramadol for Reduction of Peripheral Neuropathic Pain. J. Pharm. Sci. 2023, 112, 132–137. [Google Scholar] [CrossRef]
  92. Elsherbiny, D.A.; Abdelgawad, A.M.; Shaheen, T.I.; Abdelwahed, N.A.M.; Jockenhoevel, S.; Ghazanfari, S. Thermoresponsive Nanofibers Loaded with Antimicrobial α-Aminophosphonate-o/w Emulsion Supported by Cellulose Nanocrystals for Smart Wound Care Patches. Int. J. Biol. Macromol. 2023, 233, 123655. [Google Scholar] [CrossRef]
  93. Buonvino, S.; Ciocci, M.; Nanni, F.; Cacciotti, I.; Melino, S. New Vegetable-Waste Biomaterials by Lupin Albus L. as Cellular Scaffolds for Applications in Biomedicine and Food. Biomaterials 2023, 293, 121984. [Google Scholar] [CrossRef]
  94. Ahmad, N.; Tayyeb, D.; Ali, I.; Alruwaili, N.K.; Ahmad, W.; ur Rehman, A.; Khan, A.H.; Iqbal, M.S. Development and Characterization of Hemicellulose-Based Films for Antibacterial Wound-Dressing Application. Polymers 2020, 12, 548. [Google Scholar] [CrossRef] [Green Version]
  95. Li, Y.; Yao, M.; Luo, Y.; Li, J.; Wang, Z.; Liang, C.; Qin, C.; Huang, C.; Yao, S. Polydopamine-Reinforced Hemicellulose-Based Multifunctional Flexible Hydrogels for Human Movement Sensing and Self-Powered Transdermal Drug Delivery. ACS Appl. Mater. Interfaces 2023, 15, 5883–5896. [Google Scholar] [CrossRef]
  96. Anjum, S.; Hasan, N.; Ilmi, R. Pectin-Based Nanoformulations for Therapeutic Applications. Polym. Nanosyst. 2023, 1, 405–439. [Google Scholar] [CrossRef]
  97. Phonrachom, O.; Charoensuk, P.; Kiti, K.; Saichana, N. International Journal of Biological Macromolecules Potential Use of Propolis-Loaded Quaternized Chitosan/Pectin Hydrogel Films as Wound Dressings: Preparation, Characterization, Antibacterial Evaluation, and in Vitro Healing Assay. Int. J. Biol. Macromol. 2023, 241, 124633. [Google Scholar] [CrossRef] [PubMed]
  98. Raghavendra Naveen, N.; Anitha, P.; Gowthami, B.; Goudanavar, P.; Fattepur, S. QbD Assisted Formulation Design and Optimization of Thiol Pectin Based Polyethyleneglycol and Montmorillonite (PEG/MMT) Nanocomposite Films of Neomycin Sulphate for Wound Healing. J. Drug Deliv. Sci. Technol. 2023, 82, 104348. [Google Scholar] [CrossRef]
  99. Otles, S.; Özyurt, V.H. Biotransformation in the Production of Secondary Metabolites. Stud. Nat. Prod. Chem. 2021, 68, 435–457. [Google Scholar] [CrossRef]
  100. Kuete, V. Health Effects of Alkaloids from African Medicinal Plants. In Toxicological Survey of African Medicinal Plants; Elsevier: Amsterdam, The Netherlands, 2014; pp. 611–633. ISBN 9780128004753. [Google Scholar]
  101. Mastrangelo, S.; Capozza, M.A.; Triarico, S.; Attinà, G.; Maurizi, P.; Romano, A.; Ruggiero, A. Opioid Transdermal Delivery System: A Useful Method for Pain Management in Children. Ann. Transl. Med. 2021, 9, 185. [Google Scholar] [CrossRef]
  102. Swaminathan, S.K.; Strasinger, C.; Kelchen, M.; Carr, J.; Ye, W.; Wokovich, A.; Ghosh, P.; Rajagopal, S.; Ueda, K.; Fisher, J.; et al. Determination of Rate and Extent of Scopolamine Release from Transderm Scōp® Transdermal Drug Delivery Systems in Healthy Human Adults. AAPS PharmSciTech 2020, 21, 117. [Google Scholar] [CrossRef]
  103. Shim, K.H.; Kang, M.J.; Sharma, N.; An, S.S.A. Beauty of the Beast: Anticholinergic Tropane Alkaloids in Therapeutics. Nat. Prod. Bioprospect. 2022, 12, 33. [Google Scholar] [CrossRef]
  104. Pensado, A.; Hattam, L.; White, K.A.J.; McGrogan, A.; Bunge, A.L.; Guy, R.H.; Delgado-Charro, M.B. Skin Pharmacokinetics of Transdermal Scopolamine: Measurements and Modeling. Mol. Pharm. 2021, 18, 2714–2723. [Google Scholar] [CrossRef]
  105. Westerling, D.; Hoglund, P.; Lundin, S.; Svedman, P. Transdermal Administration of Morphine to Healthy Subjects. Br. J. Clin. Pharmacol. 1994, 37, 571–576. [Google Scholar] [CrossRef]
  106. Ahn, J.S.; Lin, J.; Ogawa, S.; Yuan, C.; O’Brien, T.; Le, B.H.C.; Bothwell, A.M.; Moon, H.; Hadjiat, Y.; Ganapathi, A. Transdermal Buprenorphine and Fentanyl Patches in Cancer Pain: A Network Systematic Review. J. Pain Res. 2017, 10, 1963–1972. [Google Scholar] [CrossRef] [Green Version]
  107. Inui, N.; Kato, T.; Uchida, S.; Chida, K.; Takeuchi, K.; Kimura, T.; Watanabe, H. Novel Patch for Transdermal Administration of Morphine. J. Pain Symptom Manag. 2012, 44, 479–485. [Google Scholar] [CrossRef] [PubMed]
  108. Luo, L.; Lane, M.E. Topical and Transdermal Delivery of Caffeine. Int. J. Pharm. 2015, 490, 155–164. [Google Scholar] [CrossRef]
  109. Londzin, P.; Zamora, M.; Kąkol, B.; Taborek, A.; Folwarczna, J. Potential of Caffeine in Alzheimer’s Disease—A Review of Experimental Studies. Nutrients 2021, 13, 537. [Google Scholar] [CrossRef]
  110. Abebe, M.W.; Appiah-Ntiamoah, R.; Kim, H. Gallic Acid Modified Alginate Self-Adhesive Hydrogel for Strain Responsive Transdermal Delivery. Int. J. Biol. Macromol. 2020, 163, 147–155. [Google Scholar] [CrossRef]
  111. Aboumanei, M.H.; Mahmoud, A.F. Design and Development of a Proniosomal Transdermal Drug Delivery System of Caffeine for Management of Migraine: In Vitro Characterization, 131I-Radiolabeling and in Vivo Biodistribution Studies. Process Biochem. 2020, 97, 201–212. [Google Scholar] [CrossRef]
  112. Setyawan, E.I.; Rohman, A.; Setyowati, E.P.; Nugroho, A.K. The Combination of Simplex Lattice Design and Chemometrics in the Formulation of Green Tea Leaves as Transdermal Matrix Patch. Pharmacia 2021, 68, 275–282. [Google Scholar] [CrossRef]
  113. Vanderpluym, J.H.; Halker Singh, R.B.; Urtecho, M.; Morrow, A.S.; Nayfeh, T.; Torres Roldan, V.D.; Farah, M.H.; Hasan, B.; Saadi, S.; Shah, S.; et al. Acute Treatments for Episodic Migraine in Adults: A Systematic Review and Meta-Analysis. JAMA-J. Am. Med. Assoc. 2021, 325, 2357–2369. [Google Scholar] [CrossRef] [PubMed]
  114. Violante, C.; Lagoa, R.; Marques-da-Silva, D. Promotion of Dermal Permeation of Bioactive Compounds Using a Microneedle Device. In Proceedings of the Biosystems in Toxicology and Pharmacology—Current Challenges, Online, 8–9 September 2022; MDPI: Basel, Switzerland, 2022; p. 4. [Google Scholar]
  115. Chandran, R.; Mohd Tohit, E.R.; Stanslas, J.; Salim, N.; Tuan Mahmood, T.M. Investigation and Optimization of Hydrogel Microneedles for Transdermal Delivery of Caffeine. Tissue Eng. Part C Methods 2022, 28, 545–556. [Google Scholar] [CrossRef]
  116. Chai, S.H.; Leventhal, A.M.; Kirkpatrick, M.G.; Eisenlohr-Moul, T.A.; Rapkin, A.J.; D’Orazio, L.; Pang, R.D. Effectiveness of Transdermal Nicotine Patch in Premenopausal Female Smokers Is Moderated by Within-Subject Severity of Negative Affect and Physical Symptoms. Psychopharmacology 2020, 237, 1737–1744. [Google Scholar] [CrossRef]
  117. Niu, J.; Yuan, M.; Chen, C.; Wang, L.; Tang, Z.; Fan, Y.; Liu, X.; Ma, Y.J.; Gan, Y. Berberine-Loaded Thiolated Pluronic F127 Polymeric Micelles for Improving Skin Permeation and Retention. Int. J. Nanomed. 2020, 15, 9987–10005. [Google Scholar] [CrossRef]
  118. Cometa, S.; Bonifacio, M.A.; Licini, C.; Bellissimo, A.; Pinto, L.; Baruzzi, F.; Mattioli-Belmonte, M.; De Giglio, E. Innovative Eco-Friendly Hydrogel Film for Berberine Delivery in Skin Applications. Molecules 2021, 26, 4901. [Google Scholar] [CrossRef] [PubMed]
  119. Li, N.; Qin, Y.; Dai, D.; Wang, P.; Shi, M.; Gao, J.; Yang, J.; Xiao, W.; Song, P.; Xu, R. Transdermal Delivery of Therapeutic Compounds with Nanotechnological Approaches in Psoriasis. Front. Bioeng. Biotechnol. 2022, 9, 804415. [Google Scholar] [CrossRef] [PubMed]
  120. 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, 1–38. [Google Scholar] [CrossRef]
  121. Ameen, D.; Michniak-Kohn, B. Development and in Vitro Evaluation of Pressure Sensitive Adhesive Patch for the Transdermal Delivery of Galantamine: Effect of Penetration Enhancers and Crystallization Inhibition. Eur. J. Pharm. Biopharm. 2019, 139, 262–271. [Google Scholar] [CrossRef]
  122. Kandil, L.S.; Hanafy, A.S.; Abdelhady, S.A. Galantamine Transdermal Patch Shows Higher Tolerability over Oral Galantamine in Rheumatoid Arthritis Rat Model. Drug Dev. Ind. Pharm. 2020, 46, 996–1004. [Google Scholar] [CrossRef]
  123. Sharma, S.; Sharma, S.; Chourasia, R.; Pandey, A.; Rai, A.K.; Sahoo, D. Alzheimer’s Disease. In Naturally Occurring Chemicals Against Alzheimer’s Disease; Elsevier: Amsterdam, The Netherlands, 2021; pp. 11–28. ISBN 9780128192122. [Google Scholar]
  124. Rorabaugh, B. Ephedrine. In xPharm: The Comprehensive Pharmacology Reference; Elsevier: Amsterdam, The Netherlands, 2007; pp. 1–6. ISBN 9780080552323. [Google Scholar]
  125. Jain, S.K.; Vyas, S.P.; Dixit, V.K. Effective and Controlled Transdermal Delivery of Ephedrine. J. Control. Release 1990, 12, 257–263. [Google Scholar] [CrossRef]
  126. Samiullah; Jan, S.U.; Gul, R.; Jalaludin, S.; Asmathullah. Formulation and Evaluation of Transdermal Patches of Pseudoephedrine HCL. Int. J. Appl. Pharm. 2020, 12, 121–127. [Google Scholar] [CrossRef]
  127. Cristofoli, M.; Kung, C.P.; Hadgraft, J.; Lane, M.E.; Sil, B.C. Ion Pairs for Transdermal and Dermal Drug Delivery: A Review. Pharmaceutics 2021, 13, 909. [Google Scholar] [CrossRef]
  128. de la Rosa, L.A.; Moreno-Escamilla, J.O.; Rodrigo-García, J.; Alvarez-Parrilla, E. Phenolic Compounds. In Postharvest Physiology and Biochemistry of Fruits and Vegetables; Elsevier: Amsterdam, The Netherlands, 2019; pp. 253–271. ISBN 9780128132784. [Google Scholar]
  129. Pinto, T.; Aires, A.; Cosme, F.; Bacelar, E.; Morais, M.C.; Oliveira, I.; Ferreira-Cardoso, J.; Anjos, R.; Vilela, A.; Gonçalves, B. Bioactive (Poly)Phenols, Volatile Compounds from Vegetables, Medicinal and Aromatic Plants. Foods 2021, 10, 106. [Google Scholar] [CrossRef]
  130. Bieżanowska-Kopeć, R.; Piatkowska, E. Total Polyphenols and Antioxidant Properties of Selected Fresh and Dried Herbs and Spices. Appl. Sci. 2022, 12, 4876. [Google Scholar] [CrossRef]
  131. Ali, A.; Wu, H.; Ponnampalam, E.N.; Cottrell, J.J.; Dunshea, F.R.; Suleria, H.A.R. Comprehensive Profiling of Most Widely Used Spices for Their Phenolic Compounds through Lc-Esi-Qtof-Ms2 and Their Antioxidant Potential. Antioxidants 2021, 10, 721. [Google Scholar] [CrossRef]
  132. Zeng, Y.; Zhou, W.; Yu, J.; Zhao, L.; Wang, K.; Hu, Z.; Liu, X. By-Products of Fruit and Vegetables: Antioxidant Properties of Extractable and Non-Extractable Phenolic Compounds. Antioxidants 2023, 12, 418. [Google Scholar] [CrossRef]
  133. Gan, R.Y.; Chan, C.L.; Yang, Q.Q.; Li, H.B.; Zhang, D.; Ge, Y.Y.; Gunaratne, A.; Ge, J.; Corke, H. Bioactive Compounds and Beneficial Functions of Sprouted Grains; Elsevier Inc.: Amsterdam, The Netherlands, 2018; ISBN 9780128115251. [Google Scholar]
  134. Zhang, Z.; Li, W.; Chang, D.; Wei, Z.; Wang, E.; Yu, J.; Xu, Y.; Que, Y.; Chen, Y.; Fan, C.; et al. A Combination Therapy for Androgenic Alopecia Based on Quercetin and Zinc/Copper Dual-Doped Mesoporous Silica Nanocomposite Microneedle Patch. Bioact. Mater. 2023, 24, 81–95. [Google Scholar] [CrossRef] [PubMed]
  135. Nisar, A. Medicinal Plants and Phenolic Compounds; IntechOpen: London, UK, 2022; Volume 11, p. 13. ISBN 0000957720. [Google Scholar]
  136. 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] [PubMed]
  137. Singh, N.; Yadav, S.S. A Review on Health Benefits of Phenolics Derived from Dietary Spices. Curr. Res. Food Sci. 2022, 5, 1508–1523. [Google Scholar] [CrossRef]
  138. Rahman, M.; Rahaman, S.; Islam, R.; Rahman, F.; Mithi, F.M.; Alqahtani, T.; Almikhlafi, M.A.; Alghamdi, S.Q.; Alruwaili, A.S.; Hossain, S.; et al. Role of Phenolic Compounds in Human Disease: Current. Molecules 2022, 27, 233. [Google Scholar] [CrossRef] [PubMed]
  139. Rathod, N.B.; Elabed, N.; Punia, S.; Ozogul, F.; Kim, S.-K.; Rocha, J.M. Recent Developments in Polyphenol Applications on Human Health: A Review with Current Knowledge. Plants 2023, 12, 1217. [Google Scholar] [CrossRef] [PubMed]
  140. Bié, J.; Sepodes, B.; Fernandes, P.C.B.; Ribeiro, M.H.L. Polyphenols in Health and Disease: Gut Microbiota, Bioaccessibility, and Bioavailability. Compounds 2023, 3, 40–72. [Google Scholar] [CrossRef]
  141. Cosme, P.; Rodríguez, A.B.; Espino, J.; Garrido, M. Plant Phenolics: Bioavailability as a Key Determinant of Their Potential Health-Promoting Applications. Antioxidants 2020, 9, 1263. [Google Scholar] [CrossRef]
  142. Nurzyńska-Wierdak, R. Phenolic Compounds from New Natural Sources—Plant Genotype and Ontogenetic Variation. Molecules 2023, 28, 1731. [Google Scholar] [CrossRef]
  143. Butkeviciute, A.; Ramanauskiene, K.; Kurapkiene, V.; Janulis, V. Dermal Penetration Studies of Potential Phenolic Compounds Ex Vivo and Their Antioxidant Activity In Vitro. Plants 2022, 11, 1901. [Google Scholar] [CrossRef] [PubMed]
  144. Gao, Q.; Li, Y.; Li, Y.; Zhang, Z.; Liang, Y. Antioxidant and Prooxidant Activities of Phenolic Acids Commonly Existed in Vegetablesand Their Relationship with Structures. Food Sci. Technol. 2022, 42, e07622. [Google Scholar] [CrossRef]
  145. Zhang, Y.; Cai, P.; Cheng, G.; Zhang, Y. A Brief Review of Phenolic Compounds Identified from Plants: Their Extraction, Analysis, and Biological Activity. Nat. Prod. Commun. 2022, 17, 1934578X211069721. [Google Scholar] [CrossRef]
  146. Luque, G.C.; Moya, M.; Picchio, M.L.; Bagnarello, V.; Valerio, I.; Bolaños, J.; Vethencourt, M.; Gamboa, S.; Tomé, L.C.; Minari, R.J.; et al. Polyphenol Iongel Patches with Antimicrobial, Antioxidant and Anti-Inflammatory Properties. Polymers 2023, 15, 1076. [Google Scholar] [CrossRef]
  147. Santi, T.D.; Siregar, T.N.; Sutriana, A.; Andini, R.; Candra, A. Phytochemical Test and Optimization of Transdermal Patches of Carica Papaya Extract: Formulation Design of Candidate Drug for Wound Healing. Biodiversitas 2022, 23, 2904–2913. [Google Scholar] [CrossRef]
  148. Hallan, S.S.; Sguizzato, M.; Mariani, P.; Cortesi, R.; Huang, N.; Simelière, F.; Marchetti, N.; Drechsler, M.; Ruzgas, T.; Esposito, E. Design and Characterization of Ethosomes for Transdermal Delivery of Caffeic Acid. Pharmaceutics 2020, 12, 740. [Google Scholar] [CrossRef]
  149. Viqhi, A.V.; Manggau, M.A.; Sartini, S.; Wahyudin, E.; Rahman, L.; Yulianti, R.; Permana, A.D.; Awal, S.A. Development of Propolis (Apis Trigona)-Loaded Nanoemulgel for Improved Skin Penetration of Caffeic Acid: The Effect of Variation of Oleic Acid Concentration. Open Access Maced. J. Med. Sci. 2021, 9, 1264–1278. [Google Scholar] [CrossRef]
  150. Contardi, M.; Lenzuni, M.; Fiorentini, F.; Summa, M.; Bertorelli, R.; Suarato, G.; Athanassiou, A. Hydroxycinnamic Acids and Derivatives Formulations for Skin Damages and Disorders: A Review. Pharmaceutics 2021, 13, 999. [Google Scholar] [CrossRef]
  151. Hendriati, L.; Fredericktho, F.F.; Hamid, I.S.; Widodo, T.; Kuncorojakti, S. The Antigangrene Activity of Transdermal Patch of Insulin Leaves (Smallanthus Sonchifolius) to Diabetic Gangrene on White Rats. J. Farm. Galen. (Galenika J. Pharmacy) 2022, 9, 30–40. [Google Scholar] [CrossRef]
  152. Zvezdin, V.; Peno-Mazzarino, L.; Radionov, N.; Kasatkina, T.; Kasatkin, I. Microneedle Patch Based on Dissolving, Detachable Microneedle Technology for Improved Skin Quality–Part 1: Ex Vivo Safety Evaluation. Int. J. Cosmet. Sci. 2020, 42, 369–376. [Google Scholar] [CrossRef]
  153. Algahtani, M.S.; Ahmad, M.Z.; Nourein, I.H.; Albarqi, H.A.; Alyami, H.S.; Alyami, M.H.; Alqahtani, A.A.; Alasiri, A.; Algahtani, T.S.; Mohammed, A.A.; et al. Preparation and Characterization of Curcumin Nanoemulgel Utilizing Ultrasonication Technique for Wound Healing: In Vitro, Ex Vivo, and In Vivo Evaluation. Gels 2021, 7, 213. [Google Scholar] [CrossRef] [PubMed]
  154. Kriplani, P.; Guarve, K.; Singh Baghel, U. Formulation Optimization and Characterization of Transdermal Film of Curcumin by Response Surface Methodology. Chin. Herb. Med. 2021, 13, 274–285. [Google Scholar] [CrossRef] [PubMed]
  155. Eckert, R.W.; Wiemann, S.; Keck, C.M. Improved Dermal and Transdermal Delivery of Curcumin with Smartfilms and Nanocrystals. Molecules 2021, 26, 1633. [Google Scholar] [CrossRef]
  156. Alven, S.; Nqoro, X.; Aderibigbe, B.A. Polymer-Based Materials Loaded with Curcumin for Wound Healing Applications. Polymers 2020, 12, 2286. [Google Scholar] [CrossRef] [PubMed]
  157. Prabhu, A.; Jose, J.; Kumar, L.; Salwa, S.; Vijay Kumar, M.; Nabavi, S.M. Transdermal Delivery of Curcumin-Loaded Solid Lipid Nanoparticles as Microneedle Patch: An In Vitro and In Vivo Study. AAPS PharmSciTech 2022, 23, 49. [Google Scholar] [CrossRef] [PubMed]
  158. Madamsetty, V.S.; Vazifehdoost, M.; Alhashemi, S.H.; Davoudi, H.; Zarrabi, A.; Dehshahri, A.; Fekri, H.S.; Mohammadinejad, R.; Thakur, V.K. Next-Generation Hydrogels as Biomaterials for Biomedical Applications: Exploring the Role of Curcumin. ACS Omega 2023, 8, 8960–8976. [Google Scholar] [CrossRef] [PubMed]
  159. Akanda, M.; Getti, G.; Douroumis, D. In Vivo Evaluation of Nanostructured Lipid Carrier Systems (NLCs) in Mice Bearing Prostate Cancer Tumours. Drug Deliv. Transl. Res. 2021, 13, 2083–2095. [Google Scholar] [CrossRef]
  160. Altamimi, M.A.; Hussain, A.; Mahdi, W.A.; Imam, S.S.; Alshammari, M.A.; Alshehri, S.; Khan, M.R. Mechanistic Insights into Luteolin-Loaded Elastic Liposomes for Transdermal Delivery: HSPiP Predictive Parameters and Instrument-Based Evidence. ACS Omega 2022, 7, 48202–48214. [Google Scholar] [CrossRef]
  161. Sabir, F.; Qindeel, M.; Rehman, A.U.; Ahmad, N.M.; Khan, G.M.; Csoka, I.; Ahmed, N. An Efficient Approach for Development and Optimisation of Curcumin-Loaded Solid Lipid Nanoparticles’ Patch for Transdermal Delivery. J. Microencapsul. 2021, 38, 233–248. [Google Scholar] [CrossRef]
  162. Zálešák, F.; Bon, D.J.Y.D.; Pospíšil, J. Lignans and Neolignans: Plant Secondary Metabolites as a Reservoir of Biologically Active Substances. Pharmacol. Res. 2019, 146, 104284. [Google Scholar] [CrossRef]
  163. Jang, W.Y.; Kim, M.Y.; Cho, J.Y. Antioxidant, Anti-Inflammatory, Anti-Menopausal, and Anti-Cancer Effects of Lignans and Their Metabolites. Int. J. Mol. Sci. 2022, 23, 15482. [Google Scholar] [CrossRef] [PubMed]
  164. Rybczyńska-Tkaczyk, K.; Grenda, A.; Jakubczyk, A.; Kiersnowska, K.; Bik-Małodzińska, M. Natural Compounds with Antimicrobial Properties in Cosmetics. Pathogens 2023, 12, 320. [Google Scholar] [CrossRef] [PubMed]
  165. Runeberg, P.; Ryabukhin, D.; Lagerquist, L.; Rahkila, J.; Eklund, P. Transformations and Antioxidative Activities of Lignans and Stilbenes at High Temperatures. Food Chem. 2023, 404, 134641. [Google Scholar] [CrossRef] [PubMed]
  166. Deresa, E.M.; Diriba, T.F. Phytochemicals as Alternative Fungicides for Controlling Plant Diseases: A Comprehensive Review of Their Efficacy, Commercial Representatives, Advantages, Challenges for Adoption, and Possible Solutions. Heliyon 2023, 9, e13810. [Google Scholar] [CrossRef]
  167. Park, R.; Park, E.J.; Cho, Y.Y.; Lee, J.Y.; Kang, H.C.; Song, I.S.; Lee, H.S. Tetrahydrofurofuranoid Lignans, Eudesmin, Fargesin, Epimagnolin a, Magnolin, and Yangambin Inhibit UDP-Glucuronosyltransferase 1A1 and 1A3 Activities in Human Liver Microsomes. Pharmaceutics 2021, 13, 3–15. [Google Scholar] [CrossRef]
  168. Sangiorgio, P.; Errico, S.; Verardi, A.; Moliterni, S.; Tamasi, G.; Rossi, C.; Balducchi, R. Bioactive Lignans from Flaxseed: Biological Properties and Patented Recovery Technologies. Nutraceuticals 2023, 3, 58–74. [Google Scholar] [CrossRef]
  169. Yeung, A.W.K.; Tzvetkov, N.T.; Balacheva, A.A.; Georgieva, M.G.; Gan, R.Y.; Jozwik, A.; Pyzel, B.; Horbańczuk, J.O.; Novellino, E.; Durazzo, A.; et al. Lignans: Quantitative Analysis of the Research Literature. Front. Pharmacol. 2020, 11, 37. [Google Scholar] [CrossRef] [Green Version]
  170. Torrisi, C.; Cardullo, N.; Russo, S.; La Mantia, A.; Acquaviva, R.; Muccilli, V.; Castelli, F.; Sarpietro, M.G. Benzo[k,l]Xanthene Lignan-Loaded Solid Lipid Nanoparticles for Topical Application: A Preliminary Study. Molecules 2022, 27, 5887. [Google Scholar] [CrossRef]
  171. Naik, N.J.; Abhyankar, I.; Darne, P.; Prabhune, A.; Madhusudhan, B. Sustained Transdermal Release of Lignans Facilitated by Sophorolipid Based Transferosomal Hydrogel for Cosmetic Application. Int. J. Curr. Microbiol. Appl. Sci. 2019, 8, 1783–1791. [Google Scholar] [CrossRef]
  172. Tasneem, R.; Khan, H.M.S.; Rasool, F.; Khan, K.U.R.; Umair, M.; Esatbeyoglu, T.; Korma, S.A. Development of Phytocosmeceutical Microemulgel Containing Flaxseed Extract and Its In Vitro and In Vivo Characterization. Pharmaceutics 2022, 14, 1656. [Google Scholar] [CrossRef]
  173. González-Sarrías, A.; Espín-Aguilar, J.C.; Romero-Reyes, S.; Puigcerver, J.; Alajarín, M.; Berná, J.; Selma, M.V.; Espín, J.C. Main Determinants Affecting the Antiproliferative Activity of Stilbenes and Their Gut Microbiota Metabolites in Colon Cancer Cells: A Structure–Activity Relationship Study. Int. J. Mol. Sci. 2022, 23, 15102. [Google Scholar] [CrossRef]
  174. Choiri, S.; Fitriastuti, R.; Faradiva, F.Z.; Rahayu, W.V. Antioxidant Activity and Nano Delivery of the Most Frequently Applied Stilbene Derivates: A Brief and Recent Review. Pharm. Sci. 2022, 28, 365–375. [Google Scholar] [CrossRef]
  175. Cebrián, R.; Li, Q.; Peñalver, P.; Belmonte-Reche, E.; Andrés-Bilbao, M.; Lucas, R.; De Paz, M.V.; Kuipers, O.P.; Morales, J.C. Chemically Tuning Resveratrol for the Effective Killing of Gram-Positive Pathogens. J. Nat. Prod. 2022, 85, 1459–1473. [Google Scholar] [CrossRef]
  176. Mattio, L.M.; Catinella, G.; Pinto, A.; Dallavalle, S. Natural and Nature-Inspired Stilbenoids as Antiviral Agents. Eur. J. Med. Chem. 2020, 202, 112541. [Google Scholar] [CrossRef]
  177. Aung, N.N.; Ngawhirunpat, T.; Rojanarata, T.; Patrojanasophon, P.; Opanasopit, P.; Pamornpathomkul, B. Enhancement of Transdermal Delivery of Resveratrol Using Eudragit and Polyvinyl Pyrrolidone-Based Dissolving Microneedle Patches. J. Drug Deliv. Sci. Technol. 2021, 61, 102284. [Google Scholar] [CrossRef]
  178. Uchida, N.; Yanagi, M.; Shimoda, K.; Hamada, H. Transdermal Delivery of Small-Sized Resveratrol Nanoparticles to Epidermis Using Anionic Phospholipids. Nat. Prod. Commun. 2020, 15, 1934578X20951443. [Google Scholar] [CrossRef]
  179. Zoabi, A.; Touitou, E.; Margulis, K. Recent Advances in Nanomaterials for Dermal and Transdermal Applications. Colloids Interfaces 2021, 5, 18. [Google Scholar] [CrossRef]
  180. Lafarge, E.; Villette, S.; Cario-André, M.; Lecomte, S.; Faure, C. Transdermal Diffusion of Resveratrol by Multilamellar Liposomes: Effect of Encapsulation on Its Stability. J. Drug Deliv. Sci. Technol. 2022, 76, 103742. [Google Scholar] [CrossRef]
  181. Kozłowska, A.; Szostak-Węgierek, D. Flavonoids–Food Sources, Health Benefits, and Mechanisms Involved. In Bioactive Molecules in Food; Springer: Cham, Switzerland, 2018; pp. 1–27. ISBN 9783319780306. [Google Scholar]
  182. Ramesh, P.; Jagadeesan, R.; Sekaran, S.; Dhanasekaran, A.; Vimalraj, S. Flavonoids: Classification, Function, and Molecular Mechanisms Involved in Bone Remodelling. Front. Endocrinol. 2021, 12, 779638. [Google Scholar] [CrossRef] [PubMed]
  183. Pecorini, G.; Ferraro, E.; Puppi, D. Polymeric Systems for the Controlled Release of Flavonoids. Pharmaceutics 2023, 15, 628. [Google Scholar] [CrossRef] [PubMed]
  184. Chiu, Y.H.; Wu, Y.W.; Hung, J.I.; Chen, M.C. Epigallocatechin Gallate/L-Ascorbic Acid–Loaded Poly-γ-Glutamate Microneedles with Antioxidant, Anti-Inflammatory, and Immunomodulatory Effects for the Treatment of Atopic Dermatitis. Acta Biomater. 2021, 130, 223–233. [Google Scholar] [CrossRef]
  185. Reddy, V.N.; Nyamathulla, S.; Pahirulzaman, K.A.K.; Mokhtar, S.I.; Giribabu, N.; Pasupuleti, V.R. Gallocatechin-Silver Nanoparticles Embedded in Cotton Gauze Patches Accelerated Wound Healing in Diabetic Rats by Promoting Proliferation and Inhibiting Apoptosis through the Wnt/β-Catenin Signaling Pathway. PLoS ONE 2022, 17, e0268505. [Google Scholar] [CrossRef]
  186. Nagula, R.L.; Wairkar, S. Cellulose Microsponges Based Gel of Naringenin for Atopic Dermatitis: Design, Optimization, in Vitro and in Vivo Investigation. Int. J. Biol. Macromol. 2020, 164, 717–725. [Google Scholar] [CrossRef] [PubMed]
  187. Ravetti, S.; Garro, A.G.; Gaitán, A.; Murature, M.; Galiano, M.; Brignone, S.G.; Palma, S.D. Naringin: Nanotechnological Strategies for Potential Pharmaceutical Applications. Pharmaceutics 2023, 15, 863. [Google Scholar] [CrossRef] [PubMed]
  188. Hering, A.; Ochocka, J.R.; Baranska, H.; Cal, K.; Stefanowicz-Hajduk, J. Mangiferin and Hesperidin Transdermal Distribution and Permeability through the Skin from Solutions and Honeybush Extracts (Cyclopia Sp.)—A Comparison Ex Vivo Study. Molecules 2021, 26, 6547. [Google Scholar] [CrossRef] [PubMed]
  189. Kazi, M.; Alhajri, A.; Alshehri, S.M.; Elzayat, E.M.; Al Meanazel, O.T.; Shakeel, F.; Noman, O.; Altamimi, M.A.; Alanazi, F.K. Enhancing Oral Bioavailability of Apigenin Using a Bioactive Self-Nanoemulsifying Drug Delivery System (Bio-SNEDDS): In Vitro, in Vivo and Stability Evaluations. Pharmaceutics 2020, 12, 749. [Google Scholar] [CrossRef]
  190. Yoon, J.H.; Kim, M.Y.; Cho, J.Y. Apigenin: A Therapeutic Agent for Treatment of Skin Inflammatory Diseases and Cancer. Int. J. Mol. Sci. 2023, 24, 1498. [Google Scholar] [CrossRef]
  191. Xie, J.; Huang, S.; Huang, H.; Deng, X.; Yue, P.; Lin, J.; Yang, M.; Han, L.; Zhang, D.K. Advances in the Application of Natural Products and the Novel Drug Delivery Systems for Psoriasis. Front. Pharmacol. 2021, 12, 644952. [Google Scholar] [CrossRef]
  192. Altamimi, M.A.; Hussain, A.; Alrajhi, M.; Alshehri, S.; Imam, S.S.; Qamar, W. Luteolin-Loaded Elastic Liposomes for Transdermal Delivery to Control Breast Cancer: In Vitro and Ex Vivo Evaluations. Pharmaceuticals 2021, 14, 1143. [Google Scholar] [CrossRef]
  193. Costa, R.; Costa Lima, S.A.; Gameiro, P.; Reis, S. On the Development of a Cutaneous Flavonoid Delivery System: Advances and Limitations. Antioxidants 2021, 10, 1376. [Google Scholar] [CrossRef]
  194. Wang, Z.; Hu, Y.; Xue, Y.; Zhu, Z.; Wu, Y.; Zeng, Q.; Wang, Y.; Han, H.; Zhang, H.; Shen, C.; et al. Mechanism Insight on Licorice Flavonoids Release from Carbopol Hydrogels: Role of “Release Steric Hindrance” and Drug Solubility in the Release Medium. Eur. J. Pharm. Sci. 2022, 179, 106307. [Google Scholar] [CrossRef] [PubMed]
  195. Ferrara, F.; Benedusi, M.; Sguizzato, M.; Cortesi, R.; Baldisserotto, A.; Buzzi, R.; Valacchi, G.; Esposito, E. Ethosomes and Transethosomes as Cutaneous Delivery Systems for Quercetin: A Preliminary Study on Melanoma Cells. Pharmaceutics 2022, 14, 1038. [Google Scholar] [CrossRef]
  196. Kharia, A.; Singhai, A.K.; Gilhotra, R. Formualtion and Evalaution of Transdermal Patch for the Treatment of Inflammation. J. Pharm. Sci. Res. 2020, 12, 780–788. [Google Scholar]
  197. Zhang, Q.; Yang, X.; Wu, Y.; Liu, C.; Xia, H.; Cheng, X.; Cheng, Y.; Xia, Y.; Wang, Y. In Vitro Evaluation of Kaempferol-Loaded Hydrogel as PH-Sensitive Drug Delivery Systems. Polymers 2022, 14, 3205. [Google Scholar] [CrossRef] [PubMed]
  198. Vu, Q.L.; Fang, C.W.; Suhail, M.; Wu, P.C. Enhancement of the Topical Bioavailability and Skin Whitening Effect of Genistein by Using Microemulsions as Drug Delivery Carriers. Pharmaceuticals 2021, 14, 1233. [Google Scholar] [CrossRef] [PubMed]
  199. Singh, S.; Grewal, S.; Sharma, N.; Behl, T.; Gupta, S.; Anwer, M.K.; Vargas-De-La-Cruz, C.; Mohan, S.; Bungau, S.G.; Bumbu, A. Unveiling the Pharmacological and Nanotechnological Facets of Daidzein: Present State-of-the-Art and Future Perspectives. Molecules 2023, 28, 1765. [Google Scholar] [CrossRef]
  200. Dall’oglio, F.; Fabbrocini, G.; Tedeschi, A.; Donnarumma, M.; Chiodini, P.; Micali, G. Licochalcone a in Combination with Salicylic Acid as Fluid Based and Hydroxy-Complex 10% Cream for the Treatment of Mild Acne: A Multicenter Prospective Trial. Clin. Cosmet. Investig. Dermatol. 2019, 12, 961–967. [Google Scholar] [CrossRef] [Green Version]
  201. Xin, Y.; Yun, S.; Yuhe, L.; Yinxue, M.; Shurui, N.; Yue, Z.; Kunming, Q.; Weidong, L. Development of Licorice Flavonoids Loaded Microemulsion for Transdermal Delivery Using CCD-Optimal Experimental Approach: Formulation Development and Characterization. Front. Nanotechnol. 2021, 3, 103742. [Google Scholar] [CrossRef]
  202. Liu, Z.; Li, M.; Wang, S.; Huang, H.; Zhang, W. Sulfur-Containing Metabolites from Marine and Terrestrial Fungal Sources: Origin, Structures, and Bioactivities. Mar. Drugs 2022, 20, 765. [Google Scholar] [CrossRef]
  203. Ruhee, R.T.; Roberts, L.A.; Ma, S.; Suzuki, K. Organosulfur Compounds: A Review of Their Anti-Inflammatory Effects in Human Health. Front. Nutr. 2020, 7, 64. [Google Scholar] [CrossRef]
  204. Iciek, M.; Bilska-wilkosz, A.; Kozdrowicki, M.; Górny, M. Reactive Sulfur Compounds in the Fight against COVID-19. Antioxidants 2022, 11, 1053. [Google Scholar] [CrossRef] [PubMed]
  205. Osipova, V.; Polovinkina, M.; Gracheva, Y.; Shpakovsky, D.; Osipova, A.; Berberova, N. Antioxidant Activity of Some Organosulfur Compounds in Vitro. Arab. J. Chem. 2021, 14, 103068. [Google Scholar] [CrossRef]
  206. Klyushova, L.S.; Kandalintseva, N.V.; Grishanova, A.Y. Antioxidant Activity of New Sulphur- and Selenium-Containing Analogues of Potassium Phenosan against H2O2-Induced Cytotoxicity in Tumour Cells. Curr. Issues Mol. Biol. 2022, 44, 3131–3145. [Google Scholar] [CrossRef] [PubMed]
  207. Pingaew, R.; Prachayasittikul, V.; Worachartcheewan, A.; Thongnum, A.; Prachayasittikul, S.; Ruchirawat, S.; Prachayasittikul, V. Anticancer Activity and QSAR Study of Sulfur-Containing Thiourea and Sulfonamide Derivatives. Heliyon 2022, 8, e10067. [Google Scholar] [CrossRef] [PubMed]
  208. Haridevamuthu, B.; Manjunathan, T.; Wilson Alphonse, C.R.; Kumar, R.S.; Thanigaivel, S.; Chandra Kishore, S.; Sundaram, V.; Gopinath, P.; Arockiaraj, J.; Bellucci, S. Functionalized Sulfur-Containing Heterocyclic Analogs Induce Sub-G1 Arrest and Apoptotic Cell Death of Laryngeal Carcinoma In Vitro. Molecules 2023, 28, 1856. [Google Scholar] [CrossRef]
  209. Abdalla, M.A.; Mühling, K.H. Plant-Derived Sulfur Containing Natural Products Produced as a Response to Biotic and Abiotic Stresses: A Review of Their Structural Diversity and Medicinal Importance. J. Appl. Bot. Food Qual. 2019, 92, 204–215. [Google Scholar] [CrossRef]
  210. Miękus, N.; Marszałek, K.; Podlacha, M.; Iqbal, A.; Puchalski, C.; Świergiel, A.H. Health Benefits of Plant-Derived Sulfur Compounds, Glucosinolates, and Organosulfur Compounds. Molecules 2020, 25, 3804. [Google Scholar] [CrossRef]
  211. Künstler, A.; Gullner, G.; Ádám, A.L.; Nagy, J.K.; Király, L. The Versatile Roles of Sulfur-Containing Biomolecules in Plant Defense—A Road to Disease Resistance. Plants 2020, 9, 1705. [Google Scholar] [CrossRef]
  212. Hill, C.R.; Shafaei, A.; Balmer, L.; Lewis, J.R.; Hodgson, J.M.; Millar, A.H.; Blekkenhorst, L.C. Sulfur Compounds: From Plants to Humans and Their Role in Chronic Disease Prevention. Crit. Rev. Food Sci. Nutr. Online ahead of print. 2022. [Google Scholar] [CrossRef]
  213. Khabibrakhmanova, A.M.; Faizova, R.G.; Lodochnikova, O.A.; Zamalieva, R.R.; Latypova, L.Z.; Trizna, E.Y.; Porfiryev, A.G.; Tanaka, K.; Sachenkov, O.A.; Kayumov, A.R.; et al. The Novel Chiral 2(5H)-Furanone Sulfones Possessing Terpene Moiety: Synthesis and Biological Activity. Molecules 2023, 28, 2543. [Google Scholar] [CrossRef]
  214. Pandey, P.; Khan, F.; Alshammari, N.; Saeed, A.; Aqil, F.; Saeed, M. Updates on the Anticancer Potential of Garlic Organosulfur Compounds and Their Nanoformulations: Plant Therapeutics in Cancer Management. Front. Pharmacol. 2023, 14, 1154034. [Google Scholar] [CrossRef] [PubMed]
  215. Perveen, S. Introductory Chapter: Terpenes and Terpenoids. In Terpenes and Terpenoids; IntechOpen: London, UK, 2018; Volume 11, p. 13. ISBN 0000957720. [Google Scholar]
  216. Çalışkan, U.K.; Karakuş, M.M. Essential Oils as Skin Permeation Boosters and Their Predicted Effect Mechanisms. J. Dermatol. Ski. Sci. 2020, 2, 24–30. [Google Scholar]
  217. Bundrla, D.S.; Kumar, H.; Pathak, A. A Review on Natural Permeation Enhancer for Transdermal Drug Delivery System and Permeation Evaluation. Int. J. Pharm. Pharm. Res. 2020, 19, 5–9. [Google Scholar]
  218. Alhasso, B.; Ghori, M.U.; Conway, B.R. Systematic Review on the Effectiveness of Essential and Carrier Oils as Skin Penetration Enhancers in Pharmaceutical Formulations. Sci. Pharm. 2022, 90, 14. [Google Scholar] [CrossRef]
  219. Hasan, A.; Farooqui, H. A Review on Role of Essential Oil as Penetration Enhancer in Transdermal Drug Delivery System. Sys. Rev. Pharm. 2021, 12, 439–444. [Google Scholar]
  220. Karaca, N.; Demirci, B.; Gavahian, M.; Demirci, F. Enhanced Bioactivity of Rosemary, Sage, Lavender, and Chamomile Essential Oils by Fractionation, Combination, and Emulsification. ACS Omega 2023, 8, 10941–10953. [Google Scholar] [CrossRef]
  221. Demurtas, O.C.; Nicolia, A.; Diretto, G. Terpenoid Transport in Plants: How Far from the Final Picture? Plants 2023, 12, 634. [Google Scholar] [CrossRef]
  222. Hosseini, M.; Pereira, D.M. The Chemical Space of Terpenes: Insights from Data Science and AI. Pharmaceuticals 2023, 16, 202. [Google Scholar] [CrossRef]
  223. Khoshnoudi-Nia, S.; Sharif, N.; Jafari, S.M. Loading of Phenolic Compounds into Electrospun Nanofibers and Electrosprayed Nanoparticles. Trends Food Sci. Technol. 2020, 95, 59–74. [Google Scholar] [CrossRef]
  224. Mamatha, J.; Gadili, S.; Pallavi, K. Formulation and Evaluation of Zidovudine Transdermal Patch Using Permeation Enhancers. J. Young Pharm. 2020, 12, s45–s50. [Google Scholar] [CrossRef]
  225. Li, J.; Xie, Q.; Ma, R.; Li, Y.; Yuan, J.; Ren, M.; Li, H.; Wang, J.; Lu, D.; Xu, Z.; et al. Recent Progress on the Synergistic Antitumor Effect of a Borneol-Modified Nanocarrier Drug Delivery System. Front. Med. 2021, 8, 750170. [Google Scholar] [CrossRef] [PubMed]
  226. Li, Z.; Gan, Y.; Kang, T.; Zhao, Y.; Huang, T.; Chen, Y.; Liu, J.; Ke, B. Camphor Attenuates Hyperalgesia in Neuropathic Pain Models in Mice. J. Pain Res. 2023, 16, 785–795. [Google Scholar] [CrossRef] [PubMed]
  227. Carreño, H.; Stashenko, E.E.; Escobar, P. Essential Oils Distilled from Colombian Aromatic Plants and Their Constituents as Penetration Enhancers for Transdermal Drug Delivery. Molecules 2023, 28, 2872. [Google Scholar] [CrossRef] [PubMed]
  228. Krishnaiah, Y.S.R.; Nada, A. Enantioselective Penetration Enhancing Effect of Carvone on the in Vitro Transdermal Permeation of Nicorandil. Pharm. Dev. Technol. 2012, 17, 574–582. [Google Scholar] [CrossRef]
  229. Isaac, M.; Holvey, C. Transdermal Patches: The Emerging Mode of Drug Delivery System in Psychiatry. Ther. Adv. Psychopharmacol. 2012, 2, 255–263. [Google Scholar] [CrossRef] [PubMed]
  230. Mishra, S.; Verma, P.; Gupta, S.; Pandey, S.; Ojha, S. Nanocarrier and Herbal Based Transdermal Patch: An Advantage Over Other Drug Delivery Systems. Ann. Ayurvedic Med. 2022, 11, 145–156. [Google Scholar] [CrossRef]
  231. Sheth, N.S.; Mistry, R.B. Formulation and Evaluation of Transdermal Patches and to Study Permeation Enhancement Effect of Eugenol. J. Appl. Pharm. Sci. 2011, 1, 96–101. [Google Scholar]
  232. Sugumar, V.; Hayyan, M.; Madhavan, P.; Wong, W.F.; Looi, C.Y. Current Development of Chemical Penetration Enhancers for Transdermal Insulin Delivery. Biomedicines 2023, 11, 664. [Google Scholar] [CrossRef]
  233. Boix-Montañés, A.; Celma-Lezcano, C.; Obach-Vidal, R.; Peraire-Guitart, C. Collaborative Permeation of Drug and Excipients in Transdermal Formulations. In Vitro Scrutiny for Ethanol:Limonene Combinations. Eur. J. Pharm. Biopharm. 2022, 181, 239–248. [Google Scholar] [CrossRef]
  234. Ogueta, I.A.; Brared Christensson, J.; Giménez-Arnau, E.; Brans, R.; Wilkinson, M.; Stingeni, L.; Foti, C.; Aerts, O.; Svedman, C.; Gonçalo, M.; et al. Limonene and Linalool Hydroperoxides Review: Pros and Cons for Routine Patch Testing. Contact Dermat. 2022, 87, 1–12. [Google Scholar] [CrossRef]
  235. Castro, E.; Dent, D. A Comparison of Transdermal Over-the-Counter Lidocaine 3.6% Menthol 1.25%, Rx Lidocaine 5% and Placebo for Back Pain and Arthritis. Pain Manag. 2017, 7, 489–498. [Google Scholar] [CrossRef] [PubMed]
  236. Zhao, H.; Ren, S.; Yang, H.; Tang, S.; Guo, C.; Liu, M.; Tao, Q.; Ming, T.; Xu, H. Peppermint Essential Oil: Its Phytochemistry, Biological Activity, Pharmacological Effect and Application. Biomed. Pharmacother. 2022, 154, 113559. [Google Scholar] [CrossRef] [PubMed]
  237. Salehi, B.; Upadhyay, S.; Orhan, I.E.; Jugran, A.K.; Jayaweera, S.L.D.; Dias, D.A.; Sharopov, F.; Taheri, Y.; Martins, N.; Baghalpour, N.; et al. Therapeutic Potential of α-and β-Pinene: A Miracle Gift of Nature. Biomolecules 2019, 9, 738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Masood, T.; Lakatos, S.; Rosta, J. Modification of the TRP Channel TRPA1 as a Relevant Factor in Migraine-Related Intracranial Hypersensitivity. Int. J. Mol. Sci. 2023, 24, 5375. [Google Scholar] [CrossRef]
  239. Bose, S.K.; Nirbhavane, P.; Batra, M.; Chhibber, S.; Harjai, K. Nanolipoidal α-Terpineol Modulates Quorum Sensing Regulated Virulence and Biofilm Formation in Pseudomonas Aeruginosa. Nanomedicine 2020, 15, 1743–1761. [Google Scholar] [CrossRef]
  240. Su, C.W.; Tighe, S.; Sheha, H.; Cheng, A.M.S.; Tseng, S.C.G. Safety and Efficacy of 4-Terpineol against Microorganisms Associated with Blepharitis and Common Ocular Diseases. BMJ Open Ophthalmol. 2018, 3, e000094. [Google Scholar] [CrossRef] [Green Version]
  241. Khan, A.; Raja, M.H.; Khan, A.; Khan, M.W.; Khan, G.M. Synthesis and Assessment of a New Tetrahydrogeraniol Derivative as Penetration Enhancer for Transdermal Drug Delivery. J. Glycom. Metab. 2016, 1, 45–53. [Google Scholar] [CrossRef] [Green Version]
  242. Silveira, Z.d.S.; Macêdo, N.S.; dos Santos, J.F.S.; de Freitas, T.S.; Barbosa, C.R.d.S.; Júnior, D.L.d.S.; Muniz, D.F.; de Oliveira, L.C.C.; Júnior, J.P.S.; da Cunha, F.A.B.; et al. Evaluation of the Antibacterial Activity and Efflux Pump Reversal of Thymol and Carvacrol against Staphylococcus Aureus and Their Toxicity in Drosophila Melanogaster. Molecules 2020, 25, 2103. [Google Scholar] [CrossRef]
  243. Chadwick, M.; Trewin, H.; Gawthrop, F.; Wagstaff, C. Sesquiterpenoids Lactones: Benefits to Plants and People. Int. J. Mol. Sci. 2013, 14, 12780–12805. [Google Scholar] [CrossRef] [Green Version]
  244. Ludwiczuk, A.; Skalicka-Woźniak, K.; Georgiev, M.I. Terpenoids. In Pharmacognosy Fundamentals, Applications and Strategies; Academic Press: Cambridge, MA, USA, 2017; ISBN 9780128020999. [Google Scholar]
  245. Arizmendi, N.; Alam, S.B.; Azyat, K.; Makeiff, D.; Befus, A.D.; Kulka, M. The Complexity of Sesquiterpene Chemistry Dictates Its Pleiotropic Biologic Effects on Inflammation. Molecules 2022, 27, 2450. [Google Scholar] [CrossRef]
  246. Albahri, G.; Badran, A.; Hijazi, A.; Daou, A.; Baydoun, E.; Nasser, M.; Merah, O. The Therapeutic Wound Healing Bioactivities of Various Medicinal Plants. Life 2023, 13, 317. [Google Scholar] [CrossRef] [PubMed]
  247. Jeon, J.H.; Kaiser, E.E.; Waters, E.S.; Yang, X.; Lourenco, J.M.; Fagan, M.M.; Scheulin, K.M.; Sneed, S.E.; Shin, S.K.; Kinder, H.A.; et al. Tanshinone IIA-Loaded Nanoparticles and Neural Stem Cell Combination Therapy Improves Gut Homeostasis and Recovery in a Pig Ischemic Stroke Model. Sci. Rep. 2023, 13, 2520. [Google Scholar] [CrossRef] [PubMed]
  248. Zhan, Y.; Xu, X.; Luo, X.; Liu, R.; Lin, Y.; Zhao, P.; Shi, J. Preparation of Tanshinone IIA Self-Soluble Microneedles and Its Inhibition on Proliferation of Human Skin Fibroblasts. Chin. Herb. Med. 2023, 15, 251–262. [Google Scholar] [CrossRef] [PubMed]
  249. Shanmugarajan, T.S.; Selvan, N.K.; Uppuluri, V.N.V.A. Development and Characterization of Squalene-Loaded Topical Agar-Based Emulgel Scaffold: Wound Healing Potential in Full-Thickness Burn Model. Int. J. Low. Extrem. Wounds 2021, 20, 364–373. [Google Scholar] [CrossRef] [PubMed]
  250. Jamal, M.; Imam, S.S.; Aqil, M.; Amir, M.; Mir, S.R.; Mujeeb, M. Transdermal Potential and Anti-Arthritic Efficacy of Ursolic Acid from Niosomal Gel Systems. Int. Immunopharmacol. 2015, 29, 361–369. [Google Scholar] [CrossRef] [PubMed]
  251. Fonseca-Santos, B.; Araujo, G.A.; Ferreira, P.S.; Victorelli, F.D.; Pironi, A.M.; Araújo, V.H.S.; Carvalho, S.G.; Chorilli, M. Design and Characterization of Lipid-Surfactant-Based Systems for Enhancing Topical Anti-Inflammatory Activity of Ursolic Acid. Pharmaceutics 2023, 15, 366. [Google Scholar] [CrossRef]
  252. Krajewska, M.; Dopierała, K.; Prochaska, K. The Biomimetic System of Oleanolic Acid and Oleic Acid at the Air-Water Interface–Interactions in Terms of Nanotechnology-Based Drug Delivery Systems. Membranes 2022, 12, 1215. [Google Scholar] [CrossRef]
  253. Shi, Z.; Pan, S.; Wang, L.; Li, S. Topical Gel Based Nanoparticles for the Controlled Release of Oleanolic Acid: Design and in Vivo Characterization of a Cubic Liquid Crystalline Anti-Inflammatory Drug. BMC Complement. Med. Ther. 2021, 21, 224. [Google Scholar] [CrossRef]
  254. Prabahar, K.; Uthumansha, U.; Elsherbiny, N.; Qushawy, M. Enhanced Skin Permeation and Controlled Release of β-Sitosterol Using Cubosomes Encrusted with Dissolving Microneedles for the Management of Alopecia. Pharmaceuticals 2023, 16, 563. [Google Scholar] [CrossRef]
  255. Afzal, O.; Akhter, M.H.; Ahmad, I.; Muzammil, K.; Dawria, A.; Zeyaullah, M.; Altamimi, A.S.A.; Khalilullah, H.; Mir Najib Ullah, S.N.; Rahman, M.A.; et al. A β–Sitosterol Encapsulated Biocompatible Alginate/Chitosan Polymer Nanocomposite for the Treatment of Breast Cancer. Pharmaceutics 2022, 14, 1711. [Google Scholar] [CrossRef]
  256. Tosato, M.G.; Orallo, D.E.; Fangio, M.F.; Diz, V.; Dicelio, L.E.; Churio, M.S. Nanomaterials and Natural Products for UV-Photoprotection. In Surface Chemistry of Nanobiomaterials-Applications of Nanobiomaterials; Elsevier: Amsterdam, The Netherlands, 2016; ISBN 9780323428613. [Google Scholar]
  257. de Souza Guedes, L.; Martinez, R.M.; Bou-Chacra, N.A.; Velasco, M.V.R.; Rosado, C.; Baby, A.R. An Overview on Topical Administration of Carotenoids and Coenzyme Q10 Loaded in Lipid Nanoparticles. Antioxidants 2021, 10, 1034. [Google Scholar] [CrossRef] [PubMed]
  258. Sharma, S.; Sathasivam, T.; Rawat, P.; Pushpamalar, J. Lycopene-Loaded Nanostructured Lipid Carrier from Carboxymethyl Oil Palm Empty Fruit Bunch Cellulose for Topical Administration. Carbohydr. Polym. Technol. Appl. 2021, 2, 100049. [Google Scholar] [CrossRef]
  259. Gomes, A.; Costa, A.L.R.; Sobral, P.J.D.A.; Cunha, R.L. Delivering β-Carotene from O/W Emulsion-Based Systems: Influence of Phase Ratio and Carrier Lipid Composition. Food Hydrocoll. Health 2023, 3, 100125. [Google Scholar] [CrossRef]
  260. Pezeshki, A.; Hamishehkar, H.; Ghanbarzadeh, B.; Fathollahy, I.; Keivani Nahr, F.; Khakbaz Heshmati, M.; Mohammadi, M. Nanostructured Lipid Carriers as a Favorable Delivery System for β-Carotene. Food Biosci. 2019, 27, 11–17. [Google Scholar] [CrossRef]
  261. Plyduang, T.; Sermkeaw, N. Development and Evaluation of a Hydrogel Containing Momordica Cochinchinensis Spreng Extract for Topical Applications. Braz. J. Pharm. Sci. 2022, 58, 1–14. [Google Scholar] [CrossRef]
  262. Algan, A.H.; Gungor-Ak, A.; Karatas, A. Nanoscale Delivery Systems of Lutein: An Updated Review from a Pharmaceutical Perspective. Pharmaceutics 2022, 14, 1852. [Google Scholar] [CrossRef]
  263. Akombaetwa, N.; Ilangala, A.B.; Thom, L.; Memvanga, P.B.; Witika, B.A.; Buya, A.B. Current Advances in Lipid Nanosystems Intended for Topical and Transdermal Drug Delivery Applications. Pharmaceutics 2023, 15, 656. [Google Scholar] [CrossRef]
  264. Abba, M.; Ibrahim, Z.; Chong, C.S.; Zawawi, N.A.; Kadir, M.R.A.; Yusof, A.H.M.; Razak, S.I.A. Transdermal Delivery of Crocin Using Bacterial Nanocellulose Membrane. Fibers Polym. 2019, 20, 2025–2031. [Google Scholar] [CrossRef]
  265. Suksaeree, J.; Pichayakorn, W.; Monton, C.; Sakunpak, A.; Chusut, T.; Saingam, W. Rubber Polymers for Transdermal Drug Delivery Systems. Ind. Eng. Chem. Res. 2014, 53, 507–513. [Google Scholar] [CrossRef]
  266. Syed Azhar, S.N.A.; Ashari, S.E.; Tan, J.K.; Kassim, N.K.; Hassan, M.; Zainuddin, N.; Mohamad, R.; Mat Azmi, I.D. Screening and Selection of Formulation Components of Nanostructured Lipid Carriers System for Mitragyna Speciosa (Korth). Havil Drug Delivery. Ind. Crop. Prod. 2023, 198, 116668. [Google Scholar] [CrossRef]
  267. Al-Ouqaili, M.T.S.; Saleh, R.O.; Amin, H.I.M.; Jawhar, Z.H.; Akbarizadeh, M.R.; Naderifar, M.; Issa, K.D.; Gavilán, J.C.O.; Nobre, M.A.L.; Jalil, A.T.; et al. Synthesize of Pluronic-Based Nanovesicular Formulation Loaded with Pistacia Atlantica Extract for Improved Antimicrobial Efficiency. Arab. J. Chem. 2023, 16, 104704. [Google Scholar] [CrossRef]
  268. Bustos-Salgado, P.; Andrade-Carrera, B.; Domínguez-Villegas, V.; Noé, V.; Mallandrich, M.; Colom, H.; Calpena-Campmany, A.; Garduño-Ramírez, M.L. In Vitro Approaches to Explore the Anticancer Potential of One Natural Flavanone and Four Derivatives Loaded in Biopolymeric Nanoparticles for Application in Topical Delivery Treatments. Pharmaceutics 2023, 15, 1632. [Google Scholar] [CrossRef]
  269. Chavda, V.P.; Nalla, L.V.; Balar, P.; Bezbaruah, R.; Apostolopoulos, V.; Singla, R.K.; Khadela, A.; Vora, L.; Uversky, V.N. Advanced Phytochemical-Based Nanocarrier Systems for the Treatment of Breast Cancer. Cancers 2023, 15, 1023. [Google Scholar] [CrossRef] [PubMed]
  270. Chellathurai, B.J.; Anburose, R.; Alyami, M.H.; Sellappan, M.; Bayan, M.F.; Chandrasekaran, B.; Chidambaram, K.; Rahamathulla, M. Development of a Polyherbal Topical Gel for the Treatment of Acne. Gels 2023, 9, 163. [Google Scholar] [CrossRef] [PubMed]
  271. Chuysinuan, P.; Pengsuk, C.; Lirdprapamongkol, K.; Thanyacharoen, T.; Techasakul, S.; Svasti, J.; Nooeaid, P. Turmeric Herb Extract-Incorporated Biopolymer Dressings with Beneficial Antibacterial, Antioxidant and Anti-Inflammatory Properties for Wound Healing. Polymers 2023, 15, 1090. [Google Scholar] [CrossRef]
  272. Dytrych, P.; Kejík, Z.; Hajduch, J.; Kaplánek, R.; Veselá, K.; Kučnirová, K.; Skaličková, M.; Venhauerová, A.; Hoskovec, D.; Martásek, P.; et al. Therapeutic Potential and Limitations of Curcumin as Antimetastatic Agent. Biomed. Pharmacother. 2023, 163, 114758. [Google Scholar] [CrossRef]
  273. Platon, I.V.; Ghiorghita, C.A.; Lazar, M.M.; Raschip, I.E.; Dinu, M.V. Chitosan Sponges with Instantaneous Shape Recovery and Multistrain Antibacterial Activity for Controlled Release of Plant-Derived Polyphenols. Int. J. Mol. Sci. 2023, 24, 4452. [Google Scholar] [CrossRef]
  274. Sharma, A.; Dheer, D.; Singh, I.; Puri, V.; Kumar, P. Phytoconstituent-Loaded Nanofibrous Meshes as Wound Dressings: A Concise Review. Pharmaceutics 2023, 15, 1058. [Google Scholar] [CrossRef]
  275. d’Angelo, I.; Provenzano, R.; Florio, E.; Lombardi, A.; Trama, U.; Ungaro, F.; Quaglia, F.; Miro, A. Transmucosal Delivery of the Medical Cannabis Oil via a Nanoemulsion Formulation. J. Drug Deliv. Sci. Technol. 2023, 79, 104004. [Google Scholar] [CrossRef]
  276. Diniz, L.R.L.; Calado, L.L.; Duarte, A.B.S.; de Sousa, D.P. Centella Asiatica and Its Metabolite Asiatic Acid: Wound Healing Effects and Therapeutic Potential. Metabolites 2023, 13, 276. [Google Scholar] [CrossRef]
  277. Feng, Y.; Li, B.; Hu, J.; Liu, W.; Tian, X.; Chen, Y.; Lu, S.; Liu, Y. Sustainable Use of Chrysanthemum Indicum Var. Aromaticum as Value-Added Green Materials in Microemulsion Hydrogels. ACS Sustain. Chem. Eng. 2023, 11, 3820–3831. [Google Scholar] [CrossRef]
  278. Hassan, A.S.; Hofni, A.; Abourehab, M.A.; Abdel-Rahman, I.A. Ginger Extract–Loaded Transethosomes for Effective Transdermal Permeation and Anti-Inflammation in Rat Model. Int. J. Nanomed. 2023, 18, 1259–1280. [Google Scholar] [CrossRef] [PubMed]
  279. Hayat, M.; Nawaz, A.; Chinnam, S.; Muzammal, M.; Latif, M.S.; Yasin, M.; Ashique, S.; Zengin, G.; Farid, A. Formulation Development and Optimization of Herbo Synthetic Gel: In Vitro Biological Evaluation and in Vivo Wound Healing Studies. Process Biochem. 2023, 130, 116–126. [Google Scholar] [CrossRef]
  280. Juszczak, A.M.; Jakimiuk, K.; Czarnomysy, R.; Strawa, J.W.; Zovko Končić, M.; Bielawski, K.; Tomczyk, M. Wound Healing Properties of Jasione Montana Extracts and Their Main Secondary Metabolites. Front. Pharmacol. 2022, 13, 894233. [Google Scholar] [CrossRef] [PubMed]
  281. Lam, P.L.; Gambari, R.; Ho, Y.W.; Wong, W.Y.; Hau, D.K.P.; Leung, T.W.T.; Leung, P.H.M.; Chui, C.H. Anti-Methicillin Resistance Staphylococcus Aureus and in Vitro Toxicology Evaluation of Corilagin-Loaded Gelatin/Agar Microspheres with Potential Biotextile Applications. Int. J. Biol. Macromol. 2023, 237, 123982. [Google Scholar] [CrossRef] [PubMed]
  282. Li, B.; Xiao, T.; Guo, S.; Wu, Y.; Lai, R.; Liu, Z.; Luo, W.; Xu, Y. Oxymatrine-Fatty Acid Deep Eutectic Solvents as Novel Penetration Enhancers for Transdermal Drug Delivery: Formation Mechanism and Enhancing Effect. Int. J. Pharm. 2023, 637, 122880. [Google Scholar] [CrossRef]
  283. Ferreira, L.M.d.M.C.; Bandeira, E.d.S.; Gomes, M.F.; Lynch, D.G.; Bastos, G.N.T.; Silva-Júnior, J.O.C.; Ribeiro-Costa, R.M. Polyacrylamide Hydrogel Containing Calendula Extract as a Wound Healing Bandage: In Vivo Test. Int. J. Mol. Sci. 2023, 24, 3806. [Google Scholar] [CrossRef]
  284. Neimkhum, W.; Anuchapreeda, S.; Lin, W.C.; Lue, S.C.; Lee, K.H.; Chaiyana, W. Enhancement of Stability and Dermal Delivery of Carissa Carandas Linn. Leaf Extract by Liquid Crystals. J. Drug Deliv. Sci. Technol. 2023, 82, 104258. [Google Scholar] [CrossRef]
  285. Perużyńska, M.; Nowak, A.; Birger, R.; Ossowicz-Rupniewska, P.; Konopacki, M.; Rakoczy, R.; Kucharski, Ł.; Wenelska, K.; Klimowicz, A.; Droździk, M.; et al. Anticancer Properties of Bacterial Cellulose Membrane Containing Ethanolic Extract of Epilobium Angustifolium L. Front. Bioeng. Biotechnol. 2023, 11, 1133345. [Google Scholar] [CrossRef]
  286. Pulat, S.; Subedi, L.; Pandey, P.; Bhosle, S.R.; Hur, J.S.; Shim, J.H.; Cho, S.S.; Kim, K.T.; Ha, H.H.; Kim, H.; et al. Topical Delivery of Atraric Acid Derived from Stereocaulon Japonicum with Enhanced Skin Permeation and Hair Regrowth Activity for Androgenic Alopecia. Pharmaceutics 2023, 15, 340. [Google Scholar] [CrossRef]
  287. Sotirova, Y.; Gugleva, V.; Stoeva, S.; Kolev, I.; Nikolova, R.; Marudova, M.; Nikolova, K.; Kiselova-Kaneva, Y.; Hristova, M.; Andonova, V. Bigel Formulations of Nanoencapsulated St. John’s Wort Extract—An Approach for Enhanced Wound Healing. Gels 2023, 9, 360. [Google Scholar] [CrossRef] [PubMed]
  288. Subramanian, S.; Duraipandian, C.; Alsayari, A.; Ramachawolran, G.; Wong, L.S.; Sekar, M.; Gan, S.H.; Subramaniyan, V.; Seethalakshmi, S.; Jeyabalan, S.; et al. Wound Healing Properties of a New Formulated Flavonoid-Rich Fraction from Dodonaea Viscosa Jacq. Leaves Extract. Front. Pharmacol. 2023, 14, 1096905. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Five main methods to enhance API absorption across the skin: drug–vehicle interaction, vesicles, and analogues; SC modification; energy-driven methods; and SC bypass [16]. (Pink color indicates the first generation of SC penetration devices; Yellow—the second generation of SC penetration devices/methods; Blue—the third generation of SC penetration devices/methods).
Figure 1. Five main methods to enhance API absorption across the skin: drug–vehicle interaction, vesicles, and analogues; SC modification; energy-driven methods; and SC bypass [16]. (Pink color indicates the first generation of SC penetration devices; Yellow—the second generation of SC penetration devices/methods; Blue—the third generation of SC penetration devices/methods).
Plants 12 02661 g001
Figure 2. Evolution of patches: (a1) ointment on a bandage system representation from ancient times; (a2) plaster (here, mustard and belladonna for controlled drug delivery, medical device developed prior to 1900s); (a3) delivery device (here, first recognized topical delivery system); (b) different types of patches; (c) different types of microneedles (minimally invasive patches). Adapted with permission [22,23]. (The arrows represent the application/removal method of the microneedle device).
Figure 2. Evolution of patches: (a1) ointment on a bandage system representation from ancient times; (a2) plaster (here, mustard and belladonna for controlled drug delivery, medical device developed prior to 1900s); (a3) delivery device (here, first recognized topical delivery system); (b) different types of patches; (c) different types of microneedles (minimally invasive patches). Adapted with permission [22,23]. (The arrows represent the application/removal method of the microneedle device).
Plants 12 02661 g002
Table 1. Mucilage-based products for WD and API delivery applications.
Table 1. Mucilage-based products for WD and API delivery applications.
Mucilage Plant SourceOther ConstituentsActive Ingredient/DrugApplicability and PropertiesRef.
Ficus reticuleta fruitGlycerol
Propylene Glycol
Span-80
Propyl/Methyl paraben
Diltiazem HClTherapy of hypertension,
angina pectoris, arrhythmia
Controlled release matrix
[40]
Ficus carica fruitPropylene glycol
Span-80
Propyl/Methyl paraben
Pioglitazone HClType II diabetes therapy
Matrix-moderated transdermal patch
Retardation of release
[41]
Povidone
Propylene glycol
Span-80
Propyl/Methyl paraben
Tramadol HClPatch for therapy of pain, inflammation, and arthritis
Sustained drug release
[42]
Artocarpus heterophyllus fruitGelatin
Glycerol
Dimethyl sulphoxide
Naproxen sodiumTherapy for rheumatoid arthritis, osteoarthritis, and ankylosis spondylitis
Sustained drug release
[43]
Ficus benghalensis fruitEudragit L
Propylene glycol
Span-60
AceclofenacTreatment of pain and inflammation
Controlled discharge matrix transdermal patches
[44]
Ficus auriculata fruitHydroxypropyl Methyl
Cellulose K4M
Polyethylene glycol-400
Tween-80
Diclofenac PotassiumTreatment of arthritis symptoms
Matrix-moderated transdermal patch
Controlled release diffusion
[45]
Ocimum basilicum seedsPolyvinyl alcohol
Glutaraldehyde
Glycerol
Tetracycline HydrochlorideWound dressing in chronic wound management
pH-sensitive cross-linked hydrogel
Drug release control
[46]
Borax
Glycerol
Zinc oxide nanoparticlesWound dressing
Crosslinked antimicrobial sponge
[47]
L-cysteine-modified alginate
Dopamine-modified hyaluronic acid
Thiolated alginate
Polydopamine
NystatinOral fungal infection treatment
Bi- and tri-layer mucoadhesive sublingual films
Controlled release
[48]
Cydonia oblonga seedsChitosan
Poly-caprolactone
Polyethylene glycol
Skin-tissue-engineered hybrid scaffolds
Dressing for continuous absorption of
wound exudates
Smart/stimuli-responsive bio-matrix
[49]
Bacterial cellulose Composite scaffold for wound dressings[50]
Leaves of Cocculus hirsutusPolyvinyl alcohol Dermal wound patches
Biodegradable biopolymer
[51]
Salvia hispanica seeds Dermatological and cosmetic formulations
Biopolymer with mucoadhesive properties
Controlled release of bioactive compounds and drugs
[52]
Lallemantia royleana
seeds
Chitosan
Chitin
Ag nanoparticles
Ciprofloxacin
Wound dressings
Controlled drug release
[53]
Althea rosea plant Functional and nutraceutical in inflammatory disorders[54]
Aloe vera leaves (fresh or powdered)Sodium alginate
Glycerol
Improved film thermal stability and transparency
Good UV light barrier
Suitable for wound healing
and drug delivery
[55]
Hydroxypropyl methylcellulose
PEG 400
CaptoprilImproved drug transdermal penetration[56]
Xanthan gumAllantoin
Natural BHA salicylic acid
Anti-inflammatory, wound epithelization, and skin regeneration hydrogel[57]
Table 2. Plant-SM-based products for WD and API delivery applications.
Table 2. Plant-SM-based products for WD and API delivery applications.
SM Plant SourceMain SMTDSApplicability and PropertiesRef.
Mitragyna Speciosa leaves’ extractAlkaloids
Flavonoids
Saponins
Lipidic carriers in oleic acid, Tween 80For topical application/wound dressing[266]
Pistacia atlantica
fruit extract
β–sitosterolLipidic carriers: span 40, pluronic F127, Cholesterol and GlycerolAntimicrobial in topical application[267]
Eysenhardtia platycarpaFlavoneBiopolymeric nanoparticlesTopical application for cancer treatment[268]
Lippia origanoides, Turnera diffusaEugenol
Carvacrol
Limonene
Carbopol hydrogelCaffeine’s skin permeation for inflammatory diseases[227]
Catharanthus roseusVinblastine
Vincristine
(alkaloids)
Lipid nanocarrierTopical treatment of breast cancer[269]
Mucuna pruritaL-Dopa
Rauvolfia serpentineReserpine
Taxus brevifoliaPaclitaxel
Vigna radiataFlavonoids
Phenolic acids
Aloe vera hydrogelTopical treatment of acne vulgaris[270]
Curcuma longaCurcuminLoaded in biopolymers
(CMC-silk sericin)
Antibacterial, antioxidant, and anti-inflammatory for wound healing[271]
NanoparticlesDifferent types of cancer[272]
Chitosan sponge—cryogelWound healing[273]
Nanofibrous meshesWound healing[274]
Cannabis sativaCannabinoids
Terpenoids
Sterols
Flavonoids
Nano emulsions
(Tween 80, PEG 400)
Transmucosal delivery[275]
Centella asiaticaFlavonoids
Plant sterols
Eugenol
Pentacyclic triterpenoids
Hydroalcoholic extractsFor topical application/WD[276]
Nanofibrous meshes[274]
Chrysanthemum indicum var. aromaticumTerpenoids
Flavonoids
Phenols
Gel formulated with Tween-80, transcutol-P, sodium hyaluronate, and glycerolTopical application for inflammatory skin disease, cosmetic use[277]
Zingiber officinaleSesquiterpene
Monoterpene hydrocarbons
NanocarriersMajor inflammatory disease[278]
Sideroxylon mascatenseBerberine
Palmatine
Jatrorrhizine
Saponins
Nanoemulsion gelWound healing[279]
Jasione montanaLuteolin
Luteolin 7-O-glucoside
Luteolin 7-O-sambubioside
Direct application (scratch cells)Wound healing[280]
Caesalpinia coriariaCorilagin (ellagitannin)Gelatin/agar microsphereWound healing[281]
Genus SophoraOxymatrine natural alkaloidDeep eutectic solvent for TD
Capric acid, Lauric acid, Myristic acid, Palmitic acid, and Stearic acid
Anti-pruritic and anti-inflammatory effects[282]
Calendula officinalisCarotenoids
Lycopene
Phenolic acids
Hydroxycinnamic acids
Flavonoids (rutin)
Coumarins (esculetin)
Polyacrylamide hydrogel containing calendula extractTherapeutic applications, which include tissue re-epithelialization and general wound healing action[283]
Carissa carandas leaf extractUrsolic acid
Phenolic and flavonoid compounds
Liquid crystals composed of 2.5% w/w coconut oil, 30% w/w Tween® 85Antioxidant, anti-inflammatory, and antiaging effects, decrease inflammatory cytokine and matrix metalloproteinase (MMP) production, as well as enhance the level of ceramide and collagen in the skin[284]
Epilobium angustifoliumPolyphenols
(flavonoids, phenolic acids, and tannins)
BiocelluloseAnticancer properties[285]
Stereocaulon japonicumAtraric acid
Phenolic compound
Volatile and nonvolatile solvents, as well as oleic acid, an effective permeation enhancerAndrogenic alopecia[286]
Azadirachta indicaAlkaloidsNanofibrous meshesWound healing[274]
Tecomella undulataAlkaloidsNanofibrous meshesWound healing
Hypericum perforatum,Polyprenylated phloroglucinol hyperforinBigel nanocarriersWound healing[287]
Dodonaea viscosa, leaf extractFlavonoids
(quercetin and kaempferol)
Direct application (scratch cells)Wound healing[288]
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

Isopencu, G.O.; Covaliu-Mierlă, C.-I.; Deleanu, I.-M. From Plants to Wound Dressing and Transdermal Delivery of Bioactive Compounds. Plants 2023, 12, 2661. https://doi.org/10.3390/plants12142661

AMA Style

Isopencu GO, Covaliu-Mierlă C-I, Deleanu I-M. From Plants to Wound Dressing and Transdermal Delivery of Bioactive Compounds. Plants. 2023; 12(14):2661. https://doi.org/10.3390/plants12142661

Chicago/Turabian Style

Isopencu, Gabriela Olimpia, Cristina-Ileana Covaliu-Mierlă, and Iuliana-Mihaela Deleanu. 2023. "From Plants to Wound Dressing and Transdermal Delivery of Bioactive Compounds" Plants 12, no. 14: 2661. https://doi.org/10.3390/plants12142661

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