Next Article in Journal
Aloe-Emodin Derivative, an Anthraquinone Compound, Attenuates Pyroptosis by Targeting NLRP3 Inflammasome in Diabetic Cardiomyopathy
Next Article in Special Issue
Chrysin Directing an Enhanced Solubility through the Formation of a Supramolecular Cyclodextrin–Calixarene Drug Delivery System: A Potential Strategy in Antifibrotic Diabetes Therapeutics
Previous Article in Journal
Antioxidant and Anti-Glycation Potential of H2 Receptor Antagonists—In Vitro Studies and a Systematic Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cyclodextrins for the Delivery of Bioactive Compounds from Natural Sources: Medicinal, Food and Cosmetics Applications

by
Stamatia Christaki
1,
Eleni Spanidi
2,
Eleni Panagiotidou
2,
Sophia Athanasopoulou
2,
Anastasia Kyriakoudi
1,
Ioannis Mourtzinos
1 and
Konstantinos Gardikis
2,*
1
Laboratory of Food Chemistry and Biochemistry, School of Agriculture, Aristotle University of Thessaloniki (AUTH), 54124 Thessaloniki, Greece
2
APIVITA SA, Industrial Park, Markopoulo, 19003 Athens, Greece
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2023, 16(9), 1274; https://doi.org/10.3390/ph16091274
Submission received: 31 July 2023 / Revised: 30 August 2023 / Accepted: 5 September 2023 / Published: 8 September 2023

Abstract

:
Cyclodextrins have gained significant and established attention as versatile carriers for the delivery of bioactive compounds derived from natural sources in various applications, including medicine, food and cosmetics. Their toroidal structure and hydrophobic cavity render them ideal candidates for encapsulating and solubilizing hydrophobic and poorly soluble compounds. Most medicinal, food and cosmetic ingredients share the challenges of hydrophobicity and degradation that can be effectively addressed by various cyclodextrin types. Though not new or novel—their first applications appeared in the market in the 1970s—their versatility has inspired numerous developments, either on the academic or industrial level. This review article provides an overview of the ever-growing applications of cyclodextrins in the delivery of bioactive compounds from natural sources and their potential application benefits.

Graphical Abstract

1. Introduction

Natural compounds have been used since ancient times for their beneficial properties and have played a crucial role in drug discovery. Such natural, biologically active compounds are derived from plants, animals or mineral sources and include, among others, vitamins, phenolic compounds, alkaloids and terpenoids. These groups of bioactive compounds possess antitumor, neuroprotective, antioxidant, anti-inflammatory and antimicrobial properties, so they have numerous applications in the medical, pharmaceutical, food and cosmetic industries [1,2,3]. However, despite their biological activities, they are characterized by poor solubility, bioavailability and stability in their bulk form, so their incorporation in various commercial health-related products is challenging. These drawbacks can be resolved through their encapsulation in delivery systems, one of which are cyclodextrins (CDs) [4].
CDs are a group of cyclic oligosaccharides produced from starch, consisting of a hydrophobic interior cavity and hydrophilic exterior. They were first described by Antoine Villiers more than one hundred years ago. In his work, Villiers reported that potato starch fermentation, under certain incubation conditions with Bacillus amylobacter, could lead to the recovery of dextrins [5,6]. However, the current term ‘cyclodextrin’ was adopted many years later and is attributed to Friedrich Cramer at the end of the 1940s [7]. Since then, an increasing number of scientific publications has been focusing on studying their structure, physicochemical properties and applications, as well as the development of new CD-based systems (e.g., nanoparticles) [8,9,10].
The most common types of CDs used in the delivery of bioactive molecules are alpha-cyclodextrin (α-CD), beta-cyclodextrin (β-CD) and gamma-cyclodextrin (γ-CD). α-CD is the smallest CD with six glucose units in the ring. It has a relatively small hydrophobic cavity and is commonly used for complexing small lipophilic molecules. β-CD is one of the most widely used CDs in bioactive compound delivery. It consists of seven glucose units in the ring, providing a larger hydrophobic cavity than α-CD. It can form inclusion complexes with a wider range of molecules including both lipophilic and hydrophilic compounds. γ-CD has eight glucose units in the ring, offering an even larger hydrophobic cavity compared to β-CD that can encapsulate larger molecules. Apart from the above three commonly used CDs, their derivatives have been introduced and applied to enhance the drug-binding capabilities of native CDs. In the first case, CDs can be chemically modified by adding methyl groups to the hydroxyl groups on the glucose units through a process called O-methylation. Examples of such CDs include heptakis (2,6-di-O-methyl)-β-cyclodextrin (DIMEB) and heptakis (2,3,6-tri-O-methyl)-β-cyclodextrin (TRIMEB). It is interesting to note that methylation affects the hydrogen bond network of CDs, thus influencing the intermolecular interactions’ strength and the host: guest flexibility. This could offer increased inclusion complex (IC) stability through stronger hydrogen bonds and/or Van der Waals interactions [11]. With another process, called acetylation, hydroxypropyl groups are added to the hydroxyl groups of CDs. So, for example, (2-hydroxy)propylated β-CD (HPβCD) can be produced from β-CD. Similarly to methylation, the degree of substitution (DS) in this case affects the CD’s properties. For example, DS-differentiated HPβCDs present various molecular recognition abilities, while their catalytic abilities also differ regarding organic reactions in water [12]. The selection of a specific CD type for the delivery of bioactive molecules depends on the physicochemical properties of the latter, the desired release profile and the intended route of administration [10].
Introducing the word “cyclodextrins” and “delivery systems,” it is evident that the interest of the scientific community toward CD research is continuously increasing (Figure 1) (Science Direct, last accessed on 28 August 2023). As shown in Figure 1, the majority of the scientific articles are related to medicinal and food applications, whereas publications that contain a combination of the keywords “cyclodextrins” and “delivery systems” and “cosmetics” are more limited. In the present review article, the incorporation of natural bioactive compounds in CDs or their derivatives along with their applications in medicine, food and cosmetics are presented and critically discussed.

2. Medicinal Applications

2.1. Legislation

CDs are exploited in medicine either as medicinal compounds or as excipients in drug formulation that confer enhanced solubility and stability properties [13]. The first medical product that took advantage of the stability effects of CDs was launched in the Japanese market in 1976 (Prostar-mon E™ sublingual tablet) [14] and, recently, at least 130 CD-formulated drugs have been listed [15]. In Japan, α-CD, β-CD and γ-CD are considered natural products and they are used in the pharmaceutical industry with few restrictions. The natural α-CD, β-CD and γ-CD are included in the GRAS (Generally Recognized as Safe) list of the Food and Drug Administration (FDA). In particular, α-CD and γ-CD have no restrictions on their consumption while there are limits on the oral intake of β-CD. In Europe, the pharmaceutical applications of CDs are regulated by the European Medicines Agency (EMA). The EMA has published a report regarding the appropriate uses of CDs in pharmaceutical products [16].

2.2. Applications

In this section, the applications of CD ICs with bioactive compounds from natural sources (e.g., polyphenols, alkaloids, terpenoids, flavonoids and essential oils) in the medical field and research are presented (Table 1). The following studies mainly focus on the improvement of in vitro activities of various bioactive compounds with potent pharmacological properties, such as anticancer, anti-inflammatory and neuroprotective activities, after their complexation with CDs.
Several compounds of natural origin have been proposed to exert antitumor activities. One example is ursolic acid, a triterpenoid found in human diet and medicinal herbs. Complexation of ursolic acid with HPβCD showed an enhanced in vitro antiproliferative activity on melanoma cell lines compared to the pure compound [17]. ICs were also efficient regarding the improvement of antitumor activities of other two terpenoids, namely, saikosaponin-d and betulinic acid. Specifically, HPβCD increased the solubility of saikosaponin-d and improved its antitumor activity in a cell line of human skin squamous cell carcinoma [18]. Regarding betulinic acid, its microencapsulation in β-CD exerted anti-proliferation effects on human breast cancer cells [19]. Similarly, HPβCD enforced the antitumor activity of the marine-derived carotenoid fucoxanthin in human colorectal carcinoma cells in vitro [20]. Camptothecin, and its natural analogue luotonin A, are alkaloids isolated from Camptotheca acuminata and are both studied for their anticancer activities. Complexation of these two alkaloids with β-CD and HPβCD enhanced their stability. Moreover, the latter ICs increased the antitumor activity of camptothecin and luotonin A compared to their free forms against a variety of cancer cell lines [21]. In another study, dihydroquercetin (taxifolin), a well-known flavonoid, was complexed with β-CD, resulting in enhanced in vitro antioxidant and antitumor activities against a hepatocellular carcinoma [22]. Similarly, β-CD also improved the cytotoxic activity of the naphthoquinone mansonone G on lung cancer cells [23]. The enhancement of biological activities of these compounds could be attributed to their complexation with CDs. Through IC formation, the bioactive compounds are protected from degradation, so their activities are prolonged. Also, their increased solubility enables their easier penetration in the human cells.
In some cases, more complicated CD-based systems are developed for the encapsulation of natural bioactive compounds. In a study, oxyresveratrol, a stilbenoid found in white mulberry, was encapsulated in a β-CD-based nanosponge. This polymer consisted of 1,1’-carbonyldiimidazole (CDI) and β-CD (β-CD: CDI 1:4) and showed a protective effect regarding oxyresveratrol’s stability during in vitro digestion, as well as stronger cell viability inhibition against prostate and colon cancer cell lines. The researchers reported that the choice of such an encapsulation system provides the ability of even slower release rates compared to simple CD complexation, so the compounds can be used in pharmaceutical products [24]. A similar system has also been used for the encapsulation of ferulic acid, leading to enhanced antiproliferation activities against breast cancer cell lines [25]. Finally, in the field of cancer research, curcumin, a widely known curcuminoid, has been encapsulated in several CD-based systems aiming to enhance its solubility and consequently its antitumor activities. Encapsulation of curcumin in HPγCD liposomes strengthened its cytotoxic activities against osteosarcoma and breast cancer cell lines [26]. Another type of curcumin combination with HPβCD and chitosan microspheres was found to improve the therapeutic index in human colorectal adenocarcinoma cell line [27]. It should be noted that the research conducted in terms of anticancer and antitumor activities of encapsulated natural bioactive compounds in CDs and their systems is quite challenging. Most of the studies report their results in vitro, so it is quite unclear whether these biological activities could be equally effective in vivo. Nevertheless, the current reported results are promising regarding the effectiveness of CD-based systems to enhance the antitumor activities of tested bioactive compounds and further research should be performed in this field.
Beyond the reinforcement of antitumor activities, complexation of bioactive compounds with CDs has been studied for various other potent therapeutic properties, such as anti-inflammatory, anti-allergic, anti-viral and others. Thymoquinone is a natural compound found in the plant Nigella sativa. Thymoquinone complexation with HPβCD in non-toxic levels ameliorated its solubility and anti-allergic effects compared to free thymoquinone or the anti-allergic drug, cetirizine [28]. Green propolis supercritical extract (GPSE) encapsulated in γ-CD exhibited anti-inflammatory activity through diet in mouse liver in vivo [29]. Known for its hepatoprotective capacity, the flavonolignan silybin (also known as silibinin), has been studied as a potent therapeutic agent for the treatment of non-alcoholic fatty liver disease (NAFLD). Silybin–HPβCD complex improved gut health though restoration of gut microbiota in high-fat diet (HFD)-fed hamsters [30]. In another study, an improved gastroprotective activity of (-)-linalool was reported in rodents upon complexation of this terpene with β-CD [31]. A recent study revealed an enhanced antiviral effect of epigallocatechin gallate, a catechin abundant in tea, against different viruses, e.g., strains of influenza virus and the coronavirus 229E. The compound formed an inclusion complex with a CD mixture composed of 0.1% β-CD, 0.4% γ-CD, and 0.2% ascorbic acid [32]. The neuroprotective effects of isothiocyanate moringin in an Alzheimer’s disease (AD) in vitro model was reported in another study. Moringin, the major bioactive compound in moringa seeds, was complexed with α-CD. Pretreatment of human neuroblastoma cell line SH-SY5Y (differentiated with retinoic acid) with the complex was able to reduce the production of proteins involved in the AD cell degeneration processes [33].
Apart from the academic research conducted in the field, several patents reflect the significance of CD-based ICs with natural bioactive compounds. In fact, the patented formulations are based on already known bioactive compounds, since they were mentioned in the paragraphs above. Indicatively, one patent refers to the improvement of curcumin’s stability through complexation with HPβCD [34], while one other describes the preparation of a bio-compatible CD complex with triterpenes, such as ursolic acid [35]. A very interesting patent deals with the complexation of betulinic acid with CD for its subsequent application in the treatment of Alzheimer’s disease [36]. Moreover, ICs of lycopene, quercetin and curcumin with HPβCD have been patented as pharmaceutical formulations for the preparation of products treating prostatitis [37]. It is clear that the interest of researchers and industry mainly focuses on bioactive compounds that possess anti-inflammatory activities so they can be used in pharmaceutical products. In addition, the compounds in the patented formulations are some of the most widely used in the literature, thus underlining their importance and justifying the interest of the scientific community towards their investigation.
Table 1. In vitro and in vivo studies of CDs complexed with natural bioactive compounds with potent medicinal applications.
Table 1. In vitro and in vivo studies of CDs complexed with natural bioactive compounds with potent medicinal applications.
CD TypeBioactive CompoundImproved CharacteristicsBiological StudyTest Subject of In Vitro/In Vivo StudyReference
HPβCDUrsolic acidStabilityAntitumor activitiesMelanoma cell lines (A375, B16 4A5 and SK-Mel 2)[17]
HPβCDSaikosaponin-dSolubility Antitumor activitiesSquamous carcinoma cell line (HSC-1)[18]
β-CDBetulinic acid-Antitumor activitiesBreast cancer cell line (MCF7)[19]
HPβCDFucoxanthinSolubility and stabilityAntitumor activitiesColorectal carcinoma (CRC) cells (HCT116 and Caco-2)[20]
β-CD and HPβCDCamptothecinStability and solubilityAntitumor activitiesBreast cancer (AREc32), lung cancer (H-23), hepatic carcinoma (HepG2), ovarian carcinoma (A2780) and neuroblastoma (SH-SY5Y) cell lines[21]
Luotonin AStability and solubility
β-CDDihydroquercetinSolubilityAntioxidant and antitumor activitiesHepatocarcinoma cell line (HepG2)[22]
β-CDMansonone GSolubility Antitumor activitiesLung cancer cells (A549)[23]
β-CD: CDI 1:4OxyresveratrolDissolution Antitumor activitiesProstate (PC-3), colon (HT-29 and HCT-116) cell lines[24]
CD-NSs Ferulic acid (FA)StabilityAntitumor activitiesBreast cancer cell lines (MCF7 and 4T1)[25]
γ-CD liposomal nanoparticlesCurcumin Solubility Antitumor activitiesOsteosarcoma (KHOS) and breast cancer (MCF-7) cell lines[26]
HPβCDThymoquinoneSolubility Antiallergic effectsRat basophilic leukemia cell line (RBL-2H3)[28]
γ-CDGreen propolis supercritical extract (GPSE)-Anti-inflammatory activitiesFemale C57BL/6NRj wild-type mice (liver)[29]
HPβCDSilybin (silibinin)Solubility Restored the gut microbiota and intestinal integrityHamsters[30]
β-CD(−)-linaloolSolubility and stability Gastroprotective effectMice[31]
β-CD, γ-CD Epigallocatechin gallateStabilityAntiviral effectInfluenza virus and HCoV-229E[32]
α-CDMoringin (MOR)-NeuroprotectionNeuroblastoma cells (SH-SY5Y) exposed to amyloid beta peptide[33]

3. Food Applications

The applications of CDs as carriers for bioactive compounds and their incorporation in various food products has been widely studied in the literature. Although CDs were initially tested and applied as drug delivery systems [8], their characteristics enabled food researchers to also widen their applications in various food matrices. Either as carriers for single bioactive compounds or as carriers for crude essential oils (EOs), CDs are effectively applied in many food products. Usually, such products are fresh, easily perishable ones (e.g., fruits, juice, fresh meat) since their composition (e.g., high moisture content) enables their microbiological and physicochemical degradation [38]. As delivery systems, CDs offer a variety of advantages in the final food product, mainly by preserving or enhancing the activities of the entrapped bioactive compounds. Firstly, the formation of inclusion complexes with CDs promotes the easier solubility of bioactive compounds in aqueous matrices [39]. This is of key importance since most of the food products that require preservation are water-based systems. In addition, encapsulation in CDs offers increased stability to the bioactive compounds as they are protected from adverse conditions (e.g., oxygen, heat) during food processing and storage. In this way, their biological activities are prolonged [40,41]. Especially for volatile compounds such as EOs and their constituents, which are most commonly applied as natural antimicrobial and antioxidant agents in foods, encapsulation in CDs decreases their evaporation rate and promotes their controlled and sustained release in the food matrix [42]. Moreover, one of the main advantages of bioactive compound encapsulation in CDs is the masking of their intense odor and taste, thus decreasing their negative effect on the organoleptic properties of foods [43].
The most widely studied and applied CD in food products is β-CD due to its affordability and compatibility with a variety of molecules [44]. β-CD is essentially odorless and white/whitish, while its aqueous solution is clear and colorless. Although β-CD is moderately soluble in water, it can easily be dissolved in warm water, whereas its solubility in ethanol is low (Regulation (EU) No 231/2012) [45]. As an approved food additive, β-CD is listed in Annexes II and III of Regulation (EC) No. 1333/2008, under the code E 459 [46]. Derivatives of β-CD, such as HPβCD, are also used. As reported by [47], CDs do not passively diffuse through biological membranes due to their hydrophilic nature, characterized by numerous hydrogen bond donors and acceptors and very low octanol–water partition coefficients. In addition, CDs present very low bioavailability when administered orally; β-CD, α-CD and their derivatives are mainly digested by bacteria in the colon, and γ-CD is completely digested in the gastrointestinal tract. In this way, they are practically nontoxic, and no CD accumulation is observed in healthy individuals with normal kidney function, even at high doses, whereas caution is recommended in the case of severely renally impaired patients [47].
Many studies in the literature have evaluated the effect of IC incorporation, between CDs and bioactive compounds, in the final food product in terms of microbiological, oxidative, color stability, and organoleptic characteristics. Guest moieties commonly studied are EOs (e.g., clove, rosemary, lemongrass) and their constituents (e.g., cuminaldehyde, thymol, citral) and phenolic compounds that are mainly hydrophobic (e.g., resveratrol, oxyresveratrol, ferulic acid, gingerols, curcumin). All the above are well-known natural compounds with important antioxidant and antimicrobial activities. Apart from direct incorporation of ICs in the food products, there are also indirect applications via active food packaging [48]. This method of preservation has been widely studied through the last years, where edible materials can be used to form a film/coating incorporating various bioactive compounds and applied externally in the food product of interest. In this way, the respective products are protected from external factors and can be preserved for a longer period of time (extension of shelf-life) [49]. Applications of active food packaging containing CD-based ICs or ICs directly incorporated in different food systems and their main effects are presented in Table 2 and Table 3, respectively.
Table 2. Applications of active packaging containing CD-based ICs in different food systems and their main effects.
Table 2. Applications of active packaging containing CD-based ICs in different food systems and their main effects.
CD TypeBioactive Compound/Guest MoietyIC 1 Preparation MethodsPackaging MaterialFood System/ModelEffects in the Final ProductReference
β-CDCinnamaldehyde (CIN)Mixing and freeze-dryingNon-woven polyethylene terephthalate (PET)Cold fresh porkPackaged pork samples with the highest tested CIN concentration were preserved for 11 days under refrigerated storage compared to control samples (7 days).[50]
Methyl-β-CDSatureja montana L. essential oil (SEO)Mixing, ultrasonication and freeze-dryingSoy soluble polysaccharide (SSPS) hydrogelMeat slicesMethyl-β-CD/SEO-SSPS hydrogel effectively reduced S. aureus counts by 3.5 log CFU/g after 7 days of storage at 4 °C.[51]
β-CDOctyl gallate (OG)Co-precipitation and freeze-dryingChitosan filmFresh fruits vegetables (blueberries and asparagus)Lower weight loss was reported in coated asparagus samples containing 0.5%, 1.0% and 2.0% β-CD/OG (3.87%, 3.12% and 2.85%, respectively), compared to control (7%) after 25 days storage at 4 °C. TVC 2 was maintained close to the initial 102–103 CFU/g in the coated asparagus samples compared to control (107 CFU/g) after 25 days of storage at 4 °C.[52]
Coated blueberries with films containing 1.0% and 2.0% β-CD/OG presented lower weight loss (2%) compared to control (7%) after 25-day storage at 4 °C. Films containing 2.0% β-CD/OG effectively preserved freshness in blueberries with a 6% rotting rate compared to control (20%).
β-CDTrans-cinnamaldehyde (TC) and citral (CI) Co-precipitation and vacuum-dryingEthylene vinyl alcohol copolymer (EVOH) filmBeefShelf-life of EVOH-β-CD-CI and EVOH-β-CD-TC coated samples was extended about 4 days at 4 °C, compared to control and coated samples without ICs. [53]
β-CDCurcumin (Cur)Mixing and freeze-dryingκ-Carrageenan (κ-Car) filmChilled porkExtension of chilled pork shelf life from 4–5 days to 10 days with application of κ-Car-β-CD-Cur film combined with light treatment, compared to pure κ-Car film and other treatments.[54]
β-CDLemongrass essential oil (LEO)Co-precipitation and dryingChitosan–gelatin (CS-Gel) coating Fresh cherry tomatoesCS/Gel coating with 7% β-CD/LEO presented high antibacterial activity against P. aurantiogriseum in cherry tomatoes artificially during 20 days of cold storage at 8 °C.[55]
α-CDBenzyl isothiocyanate (BITC)Mixing, ultrasonication and vacuum freeze-dryingChitosan (CS) film BeefCS-α-CD-BITC-coated beef samples presented lower TVC, TVB-N 3 and TBARS 4 values and higher overall acceptability score, compared to PET- 5 and CS-coated samples after 12 days of refrigerated storage. [56]
1 ICs: Inclusion complexes, 2 TVC: total viable count, 3 TVB-N: total volatile base nitrogen, 4 TBARS: thiobarbituric acid reactive substances, 5 PET: polyethylene terephthalate.
Films/coatings incorporating ICs have been applied in meat products (beef, pork) for preservation (Table 2). Zhou et al. reported the extension of refrigerated storage of fresh pork packaged with non-woven PET containing CIN-β-CD ICs by 4 more days compared to the control (non-packaged samples) [50]. Similar results were reported by Wu et al., where κ-carrageenan films containing Cur-β-CD ICs, combined with light treatment, extended the shelf-life of chilled pork by 5 days, compared to pure κ-carrageenan films (no extension) [54]. Chitosan-based films with ICs of benzyl isothiocyanate, a compound found in the plants of the Brassicaceae family, and α-CD effectively preserved beef samples after 12 days of refrigerated storage. These coated samples were less oxidized after storage (lower TVB-N and TBARS values) and more microbiologically stable (lower TVC) compared to samples coated with pure chitosan films or PET [56]. Chen et al. used films from ethylene vinyl alcohol copolymer (EVOH) incorporating ICs of β-CD with two essential oil constituents, namely, trans-cinnamaldehyde and citral, for the preservation of beef [53]. The results showed that EVOH-coated beef samples containing ICs were preserved for 4 more days in refrigerated storage compared to both non-coated samples and EVOH-coated samples without ICs. It can be concluded that encapsulation of bioactive compounds in the cavity of CDs prior to their incorporation in the film/coating-forming solution enhances their solubility and preserves their biological activities. However, for comparison reasons, it would be also useful to prepare films/coatings containing the pure compounds (without CDs). In this way, an overall conclusion can be drawn regarding not only the extension of biological activities of bioactive compounds, but also the other advantages that CDs offer (sustained release, odor masking, etc.).
Table 3. Applications of CD-based ICs in different food systems and their main effects.
Table 3. Applications of CD-based ICs in different food systems and their main effects.
CD TypeBioactive Compound/Guest MoietyIC 1 Preparation Methods Food System/ModelEffects in the Final ProductReference
β-CDCuminaldehyde (CUM)Ultrasonication, cold nitrogen plasma (CNP) treatment and freeze-dryingVegetable juices (tomato and cucumber)CNP-treated ICs decreased the E. coli O157:H7 population from 3.5 log CFU/mL to 2.51 (12 °C) and 1.29 log CFU/mL (4 °C) on cucumber juice, and to 2.58 (12 °C) and 1.33 log CFU/mL (4 °C) on tomato juice after 3 days of storage, compared to control (no added ICs).[57]
β-CDFerulic acid (FA)Crosslinking of β-CD with diphenyl carbonate (nanosponges preparation), agitation and freeze-drying Pomegranate juiceHighest TPC 2 and antioxidant activity of pomegranate juice treated with FA-CD-NSs 3 containing 500 mg/L FA was reported after 30 days of storage at 4 °C compared to control and samples containing free FA.
Total anthocyanins were better stabilized in pomegranate juice treated with FA-CD-NSs containing 250 mg/L FA after 30 days of storage at 4 °C, compared to control and samples containing free FA, through co-pigmentation effect.
[58]
β-CDClove essential oil (CEO)β-CD-metal organic frameworks (β-CD-MOFs) preparation through methanol vapor diffusion, mixing and freeze-dryingChinese bacon (preserved meat product)The lowest MDA 4 and POV 5 values were reported in Chinese bacon preserved with CEO-β-CD-MOFs in all tested concentrations, after 3 days of preservation and 15 days of fermentation compared to control, samples containing free CEO or BHT 6.[59]
β-CDFish oil (FO)Homogenization for emulsion formation and ultrasonicationYogurtFO-IC-treated yogurt presented greater syneresis reduction and lower POV values, but higher DHA 7 and EPA 8 content, after 21 days of storage at 4 °C compared to control and samples containing free FO. [60]
FO-IC-treated yogurt was significantly better accepted regarding sensory characteristics compared to the free-FO-treated one.
γ-CDResveratrol (RSV)Mixing, snap-freezing and freeze-dryingLemon juiceRSV encapsulation in γ-CD improved its solubility in lemon juice by nine times compared to free RSV (43.1% and 4.8% dissolution, respectively) at day 0. Higher RSV content was reported in γ-CD-RSV-treated lemon juice after 28 days of storage under dark conditions (room temperature or 4 °C).[61]
HPβCDApple polyphenols (AP)Mixing and freeze-dryingLambFrozen-stored lamb treated with 1.6 mg/mL AP/HPβCD-ICs presented the lowest carbonyl content (protein oxidation parameter) and improved muscle tissue structure after 40 days of storage compared to control and other tested IC concentrations. [62]
γ-CDEpigallocatechin-3-gallate (EGCG)Co-precipitation and freeze-dryingShrimp surimi productsγ-CD-EGC-treated shrimp surimi products were better preserved regarding lipid oxidation phenomena and browning effects from EGCG oxidation after 5 weeks under refrigerated storage compared to control and free-EGCG-treated samples.[63]
γ-CDGingerols (GINs)Co-precipitation and dryingYogurtLower ΔΕ in γ-CD-GIN-treated yogurt compared to the free-GIN-treated one regarding L*, a* and b* color parameters (control used as reference). [64]
γ-CD-GIN-treated yogurt presented higher ABTS radical scavenging activity compared to control and the free-GIN-treated one.
HPβCDThymol (Th)Ultrasonication and freeze-dryingTomatoesA 66.55% lower disease incidence from B. cinerea in tomato samples treated with 30 mg/mL HPβCD-Th-ICs compared to control after storage for 3 days at 25 °C. [65]
β-CD and HPβCDOxyresveratrol (Ox)Agitation and spray-drying Grape juiceOx-β-CD- and Ox-HPβCD-treated samples, combined with ascorbic acid, presented the lowest L* and ΔΕ value differences (compared to 0 h) after 24 h of storage at room temperature, indicating an anti-browning effect. [66]
β-CDRosemary essential oil (REO)Co-precipitation and dryingTomato juiceIn REO-β-CD-treated tomato juice, the population of S. pastorianus decreased from 5.5 log CFU/100 mL (day 0) to 2 log CFU/100 mL after 15-day storage at 5 °C, and this difference was significantly higher compared to control and free-REO-treated samples. [67]
1 ICs: inclusion complexes, 2 TPC: total phenolic content, 3 NSs: nanosponges, 4 MDA: malondialdehyde, 5 POV: peroxide value, 6 BHT: butylated hydroxytoluene, 7 DHA: docosahexaenoic acid, 8 EPA: eicosapentaenoic acid.
In contrast to the active food packaging containing ICs, which has been mainly applied to meat products, pure ICs containing bioactive compounds have been commonly used for juice preservation (Table 3). This is expected, since ICs are in solid, water-soluble form (powder), thus enabling their solubilization in aqueous matrices. In addition, due to the high amount of sugars and water present in juices, the incorporation of antioxidant and antimicrobial agents is crucial for their preservation during storage. CUM-β-CD ICs that were treated with cold nitrogen plasma were added in vegetable juices for evaluation of their antibacterial activity against E. coli O157: H7. The results showed that ICs were effective against E. coli, since its population decreased both in 4 °C and 12 °C compared to control samples (no added ICs). The decrease was bigger in the case of 4 °C, implying a synergy between cold storage and the antibacterial agent, cuminaldehyde [57]. Garcia-Sotelo et al. evaluated the application of REO-β-CD ICs in tomato juice regarding the antimicrobial activity against the yeast S. pastorianus [67]. The population of S. pastorianus decreased by 3.5 log CFU/100 mL in the IC-treated samples after 15 days of storage at 5 °C, whereas control and samples treated with free (non-encapsulated) REO did not present the same population decrease. In a study by Amani et al. [58], the incorporation of FA-β-CD ICs in pomegranate juice was studied in terms of anthocyanin content, among other factors. The results showed that, in the presence of ICs containing 250 mg/L FA, total anthocyanins were better stabilized in the juice after 30 days of storage at 4 °C compared to control and samples containing free FA due to a co-pigmentation effect. Oxyresveratrol, a natural stilbenoid with high antioxidant activity, was complexed with β- and HPβCD and added in grape juice for elimination of browning phenomena responsible for the product’s degradation [66]. The application of both ICs and ascorbic acid in grape juice effectively preserved the color (in terms of lightness L* and ΔΕ) after 24 h of storage at room temperature.
The importance of CDs as carriers for bioactive compounds is also highlighted by the number of patented works regarding applications of such complexes in various food systems. Indicatively, the preparation of ICs of at least one CD, or derivatives, complexed with terpene glycosides (e.g., steviol glycosides) for the production of beverages in the food industry has been patented [68]. The complexation of such glycosides with CDs is expected to increase their solubility in the beverage matrix. Another patented invention regarding non-alcoholic beverages focuses on the reduction and future replacement of conventional preserving agents in such products by suggesting an antimicrobial system based on CD complexes with natural compounds (e.g., limonene, cinnamaldehyde) that will not increase the beverage pH higher than 7.5 [69]. In another patent, the development of ICs of sucralose (sweetener) with CD is described [70]. Later on, another patented invention focused on the development of gum containing the above ICs, aiming to preserve the activity of sucralose, i.e., the sweet taste of gum, throughout chewing [71]. One more very interesting, patented invention describes the production of CD complexes with colorant-eliminating agents and their incorporation in confections (e.g., gum) in order to remove colorants from teeth surfaces [72]. The researchers suggest the incorporation of such ICs in the final stages of gum production. The preparation and preservation of a bakery product containing ICs of natamycin with CD is presented in another patented invention. For antimicrobial effectiveness, the ICs should be applied on the surface of the bakery product and the amount of natamycin should range from 0.1 pg/cm2 to 7.0 pg/cm2 [73]. Last but not least, the development of ICs between carotenoids, such as astaxanthin or lycopene, with CDs for the improvement of their storage stability compared to carotenoids in their free form is described in another patent [74]. The researchers highlight the ability of the ICs containing carotenoids to also improve pigmentation in animal tissues (e.g., salmon), and be used as feed or food supplement. It can be observed that the CD-based complexes can be used in various food products. The interest of researchers is basically focused on the delivery of bioactive compounds that can act as preservatives, while in most cases the ability of CDs to eliminate the taste, aroma or color of bioactive compounds is taken into consideration for the development of novel formulations.

4. Dietary Supplements Applications

The advantages of CDs regarding improved solubility, absorption and bioavailability of the entrapped bioactive compounds have favored their applications in dietary supplements. Similarly to food applications, CDs have been employed as carriers for sensitive, easily-degradable, lipophilic compounds that exert important biological activities and should be preserved throughout the gastrointestinal tract (GT) until their absorption in the human body [75]. An overview of literature studies regarding applications of CD-based inclusion complexes in dietary supplements is presented in Table 4. In all cases, the bioavailability of bioactive compounds of interest was increased, highlighting the ability of ICs to effectively protect and deliver them through the GT. For this reason, commercial dietary supplements containing CD complexes have already been developed.
A dietary supplement with R-alpha lipoic acid complexed with CDs (R-ALA Cyclodextrin Complex Tablets®) is used for mitochondrial function enhancement and promotion of metabolic function and antioxidant activity in a suggested dose of 500 mg/day. The complexation improves both the stability and bioavailability of R-ALA. Turkesterone, a steroid hormone of Ajuga Turkestanica, and ecdysterone, a steroid hormone from Cyanotis arachnoidea root extract (similar molecular structure to testosterone), are used as dietary supplements for improvement of athletic performance, promotion of lean muscle growth, support of the immune system and muscle repair and recovery. Complexation of these compounds with HPβCD offers maximum absorption and increased potency (Ecdysterone Complexed With Hydroxypropyl-β-Cyclodextrin®, Turkesterone Complexed With Hydroxypropyl-β-Cyclodextrin®). Dietary supplements of curcumin complexed with γ-CD (Curcumin Extrakt 45®) and manuka honey complexed with α-CD (MGOTM 400 Manuka honey®) are mainly used for their anti-inflammatory and antioxidant activities. In both cases, the absorption and bioavailability of the encapsulated bioactive compounds have increased. γ-CD was also used for the encapsulation of ubiquinone (CoQ10), an important coenzyme of the human body, in a dietary supplement that promotes the replenishment of CoQ10 levels in the body and provides energy (Co-Enzyme Q10+®). It was reported that the CoQ10 absorption was improved by 18× through complexation with γ-CD compared to free CoQ10.
Apart from commercial products, there are also patented inventions based on applications of ICs in dietary supplements. Regarding CoQ10 again, a patent describes the preparation of water-soluble ICs of the compound with β-CD that aims to increase the water solubility, bioavailability and biological activities of CoQ10 [76]. The development of a dietary supplement containing nano-ICs of carotenoids with γ-CD is pending patenting [77]. The researchers propose an easy mixing method for the dietary supplement preparation, which also includes fish oil and beeswax, apart from the ICs. In the same line, the water solubility of trans-resveratrol, an antioxidant polyphenol, is improved through complexation with β-CD [78]. The ICs are proposed as useful phytotherapeutic ingredients for incorporation in nutraceutical (pharmaceutical and/or dietary) supplements. The preparation of nutraceutical tablets containing ICs of β-CD with fatty acids (e.g., omega-3, omega-6) for oral administration has also been recently patented [79]. The respective tablets are proposed for the prevention or treatment of cardiovascular-system-, skin- or bone-related disorders.
Table 4. Applications of CD-based ICs in dietary supplements and their main effects.
Table 4. Applications of CD-based ICs in dietary supplements and their main effects.
CD TypeBioactive Compound/Guest MoietyTargeted Disease/Health ProblemEffects/Key FindingsReference
γ-CDVitamins D3 and ECystic fibrosis (with pancreatic insufficiency)Improved bioavailability of fat-soluble vitamins D3 and E[80]
HPβCDCurcumin and piperine (CUR-PIP)Diseases of infectious or neurodegenerative etiologyImproved membrane permeability, solubility and antimicrobial and antioxidant activity of CUR-PIP was reported after complexation with HPβCD. The system presented enhanced inhibitory activities against butyrylcholinesterase and acetylcholinesterase.[81]
β-CDOregano essential oil (OEO)Intestinal parasitic infectionLipid-based nutrient supplement (LNS) containing β-CD-OEO inclusion complexes (27.2 mg/20 g LNS) was stable through the gastrointestinal phases and no differences were reported in sensorial characteristics compared to control.[82]
β-CDDiosgenin (DIO)Hyperlipidemia, hyperglycemia, reduced skin thicknessHighly improved oral bioavailability of β-CD-DIO complexes and skin distribution of diosgenin compared to oral administration of free diosgenin.[83]

5. Cosmetics Applications

In the cosmetics industry, products usually appear in conventional creams, gels, lotions and solid dosage forms. The current needs and requirements of this industry have led to more intense research, development and application of new formulas based onnano-encapsulation and micro-encapsulation techniques. The difficulty of delivering bioactive ingredients to the skin, mainly due to the strong barrier of the stratum corneum (SC), requires new delivery systems to create products/formulas with superior technological performance [84,85,86].
In the early 1980s, the first review articles were already mentioning the potential application of CDs in cosmetic and personal care products. Several advantages have already been reported with the use of CDs in cosmetic products, such as the increase in solubility and the rate of solubilization, change in the viscosity or the use of more natural solvents by avoiding the use of organic solvents. In addition, CDs protect the guest bioactive components against hydrolysis or oxidation, degradation induced by heat or light, volatile ingredient loss and reactions with other cosmetic ingredients. The reduction or even elimination of undesired characteristics such as taste or odor and hygroscopicity is one of the advantages of using CDs. The effect of CDs in reducing the microbiological load was also observed since they do not constitute a nutrient medium. Furthermore, encapsulation in CDs offers improvement in handling bioactive compounds, with oily or liquid substances transformed to a solid form, and increases the stability of emulsions and thermal stability of oils [85,87,88,89]. In recent decades, the application of CDs for skin use has been particularly widespread [90] in products aimed at sun protection, wound healing, acne, psoriasis and dermatitis as well as for use in deodorants and shampoos [84].
Bioactive compounds from natural sources are used in the cosmetic industry as they most often present a better safety profile than synthetic ones [91]. Despite having scientifically documented biological activities, they face challenges with their handling and therefore their use and activity is limited. Some of the issues improved with the use of CDs are their bioavailability, solubility and stability [91].
Many bioactive compounds from natural sources have been evaluated for potential use in the cosmetic industry (Table 5). For example, three different types of CDs were used (HPβCD, M-β-CD and HPγCD) to increase the shelf life, antimicrobial properties and loading capacity of linalool for potential applications in personal care and cosmetics [92,93,94]. Encapsulation with β-CD improves the antimicrobial and antioxidant activity of Lippia graveolens essential oil for potential use in cosmetics [95]. Celastrus paniculatus seed oil was loaded in HPβCD to increase skin penetration and stability [96]. Total phenolics, phenolic acids and flavonoids from Helichrysum italicum were loaded in HPβCD, enhancing a metabolite with anti-inflammatory properties as well as anti-elastase and anti-collagenase activities, making it able to use in the cosmetic industry [97]. Also, the use of CDs has been investigated for the separation of bioactive targets for potential use in cosmetics, such as isolating glabridin from a widely used plant in cosmetics, Glycyrrhiza glabra, with the help of β-CD and SBE-β-CD [98].
There are several inventions on cyclodextrin entrapment of natural bioactive compounds for cosmetic use. Indicatively, silymarin from the plant Silybum marianum L. is entrapped in SBE-β-CDs to prevent skin aging, reduce oxidative stress in skin cells, decrease facial redness in rosacea-prone skin and increase collagen production [107]. In another patented invention, different types of CDs (α-CDs, β-CDs, γ-CDs, HPβCDs, and HPγCDs) are used to improve the solubility and stability of polyphenol mangiferin from the plant Mangifera indica L. (mango) [108]. HPβCD was used to increase the solubility of the bioactive components of propolis and increase the efficiency of the extraction, creating a more stable extract rich in antioxidants enriching face and body cosmetic products [109].
A very interesting emerging field is focused on the use of CDs as components of advanced drug delivery systems that are complemented by other particles. In such applications, CDs may be used as encapsulation efficiency enhancers and controlled-release modulators. In the following two examples, CDs were used along with liposomes in order to form a single delivery system with advanced properties. By using HPβCD along with liposomes, a stable colloidal dispersion system with loaded propolis bioactive compounds (polyphenols) was formed. A percentage of 25% up to 60% of the encapsulated propolis compounds are released within 8 h, while total release takes place within a day. The application of this system on primary human skin fibroblasts (NHDF) results in a 100% increase in viability over the control sample, representing an increase in adenosine triphosphate (ATP) therein [110]. In another patent, HPβCD was used to prepare a colloidal stabilization system and controlled release of royal jelly ingredients. CD encapsulates the polyphenols and enhances the system’s penetration into the skin, and its combination with liposomes allows the controlled release of the ingredients [111]. The combination of CD complexes and liposomes for the development of product formulations that are more easily absorbed by the skin, while retaining the biological activity of the encapsulated compounds, perhaps reflects the current market needs. In addition, the competitiveness of the cosmetic industry renders the continuous search and development of novel, natural products crucial. This fact imposes a challenge on researchers to come up with new combinations of delivery systems.
The main concern for the selection of CDs in cosmetics is their safety during their use. Studies have shown contradictory results for skin safety using CDs [112], but the only group of CDs that may affect skin barrier properties are the methylated derivatives [113,114,115], though it depends on their concentration. Generally, natural CDs have been shown to be compatible with the skin and are considered non-irritating and safe for dermal use, as they do not cause disruption of the SC [84,116,117].

6. Conclusions

CDs have emerged as remarkably versatile carriers for delivering bioactive compounds derived from natural sources in medicinal, food and cosmetic formulations. Their primary capability lies in their capacity to enhance the solubility, stability and bioavailability of these natural bioactive compounds. This attribute significantly contributes to amplifying therapeutic efficacy in medical applications, driving advancements in functional foods, and fostering innovation in cosmetic formulations. The integration of CDs within these sectors not only promises the development of advanced delivery systems but also unlocks the full range of benefits offered by bioactive compounds sourced from nature. By encapsulating these compounds, CDs facilitate sustained and targeted release, thereby magnifying their impact and expanding their potential uses. Consequently, the convergence of CDs with these industries paves the path for various delivery mechanisms, fully harnessing the multifaceted benefits provided by bioactive compounds derived from natural sources. This integration not only expands the horizons of medical treatments but also holds the potential to revolutionize dietary practices and redefine approaches to cosmetic applications. Although the research around CDs is not a novel field, their ability to be employed in various fields as well as the quite limited number of in vivo studies regarding bioavailability of encapsulated compounds, coupled with the current trend of increasing use of natural bioactive compounds, will ensure the interest around these carriers continues to grow.

Author Contributions

Conceptualization, I.M., K.G., A.K. and E.S.; writing—original draft preparation, all authors; writing—review and editing, S.C., A.K. and E.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  2. González-Manzano, S.; Dueñas, M. Applications of Natural Products in Food. Foods 2021, 10, 300. [Google Scholar] [CrossRef] [PubMed]
  3. Liu, J.-K. Natural products in cosmetics. Nat. Prod. Bioprospect. 2022, 12, 40. [Google Scholar] [CrossRef] [PubMed]
  4. Jambhekar, S.S.; Breen, P. Cyclodextrins in pharmaceutical formulations I: Structure and physicochemical properties, formation of complexes, and types of complex. Drug Discov. Today 2016, 21, 356–362. [Google Scholar] [CrossRef] [PubMed]
  5. Villiers, A. Sur la fermentation de la fecule par l’action du ferment butyrique. Compt. Rend. Acad. Sci. 1891, 112, 435–437. [Google Scholar]
  6. Villiers, A. Sur le monde d’action du ferment butyrique dans la transformation de le fecule en dextrine. Compt. Rend. Acad. Sci. 1891, 113, 144. [Google Scholar]
  7. Cramer, F. EINSCHLUSSVERBINDUNGEN VON CYCLODEXTRINEN UND DIE JOD-REAKTION DER STARKE. Angew. Chem. 1951, 63, 487. [Google Scholar]
  8. Jansook, P.; Ogawa, N.; Loftsson, T. Cyclodextrins: Structure, physicochemical properties and pharmaceutical applications. Int. J. Pharm. 2018, 535, 272–284. [Google Scholar] [CrossRef]
  9. Pandey, A. Cyclodextrin-based nanoparticles for pharmaceutical applications: A review. Environ. Chem. Lett. 2021, 19, 4297–4310. [Google Scholar] [CrossRef]
  10. Morin-Crini, N.; Fourmentin, S.; Fenyvesi, É.; Lichtfouse, E.; Torri, G.; Fourmentin, M.; Crini, G. 130 years of cyclodextrin discovery for health, food, agriculture, and the industry: A review. Environ. Chem. Lett. 2021, 19, 2581–2617. [Google Scholar] [CrossRef]
  11. Chen, H.; Ji, H. Effect of Substitution Degree of 2-Hydroxypropyl-β-Cyclodextrin on the Alkaline Hydrolysis of Cinnamaldehyde to Benzaldehyde. Supramol. Chem. 2014, 26, 796–803. [Google Scholar] [CrossRef]
  12. Geue, N.; Alcázar, J.J.; Campodónico, P.R. Influence of β-Cyclodextrin Methylation on Host-Guest Complex Stability: A Theoretical Study of Intra- and Intermolecular Interactions as Well as Host Dimer Formation. Molecules 2023, 28, 2625. [Google Scholar] [CrossRef] [PubMed]
  13. Braga, S.S. Cyclodextrins: Emerging Medicines of the New Millennium. Biomolecules 2019, 9, 801. [Google Scholar] [CrossRef] [PubMed]
  14. Uekama, K.; Hirayama, F. Inclusion Complexation of Prostaglandin F with α- and β-Cyclodextrins in Aqueous Solution. Chem. Pharm. Bull. 1978, 26, 1195–1200. [Google Scholar] [CrossRef]
  15. Puskás, I.; Szente, L.; Szőcs, L.; Fenyvesi, É. Recent List of Cyclodextrin-Containing Drug Products. Period. Polytech. Chem. Eng. 2023, 67, 11–17. [Google Scholar] [CrossRef]
  16. European Medicines Agency. Available online: https://www.ema.europa.eu/en/cyclodextrins (accessed on 25 August 2023).
  17. Oprean, C.; Mioc, M.; Csányi, E.; Ambrus, R.; Bojin, F.; Tatu, C.; Cristea, M.; Ivan, A.; Danciu, C.; Dehelean, C.; et al. Improvement of ursolic and oleanolic acids’ antitumor activity by complexation with hydrophilic cyclodextrins. Biomed. Pharmacother. 2016, 83, 1095–1104. [Google Scholar] [CrossRef]
  18. Hu, S.C.-S.; Lai, Y.-C.; Lin, C.-L.; Tzeng, W.-S.; Yen, F.-L. Inclusion complex of saikosaponin-d with hydroxypropyl-β-cyclodextrin: Improved physicochemical properties and anti-skin cancer activity. Phytomedicine 2019, 57, 174–182. [Google Scholar] [CrossRef]
  19. Sun, Y.-F.; Song, C.-K.; Viernstein, H.; Unger, F.; Liang, Z.-S. Apoptosis of human breast cancer cells induced by microencapsulated betulinic acid from sour jujube fruits through the mitochondria transduction pathway. Food Chem. 2013, 138, 1998–2007. [Google Scholar] [CrossRef]
  20. Sun, X.; Zhu, J.; Liu, C.; Wang, D.; Wang, C.-Y. Fabrication of fucoxanthin/2-hydroxypropyl-β-cyclodextrin inclusion complex assisted by ultrasound procedure to enhance aqueous solubility, stability and antitumor effect of fucoxanthin. Ultrason. Sonochem. 2022, 90, 106215. [Google Scholar] [CrossRef]
  21. González-Ruiz, V.; Cores, Á.; Martín-Cámara, O.; Orellana, K.; Cervera-Carrascón, V.; Michalska, P.; Olives, A.I.; León, R.; Martín, M.A.; Menéndez, J.C. Enhanced Stability and Bioactivity of Natural Anticancer Topoisomerase I Inhibitors through Cyclodextrin Complexation. Pharmaceutics 2021, 13, 1609. [Google Scholar] [CrossRef]
  22. Zhang, Y.; Yu, J.; Dong, X.-D.; Ji, H.-Y. Research on Characteristics, Antioxidant and Antitumor Activities of Dihydroquercetin and Its Complexes. Molecules 2017, 23, 20. [Google Scholar] [CrossRef] [PubMed]
  23. Mahalapbutr, P.; Wonganan, P.; Charoenwongpaiboon, T.; Prousoontorn, M.; Chavasiri, W.; Rungrotmongkol, T. Enhanced Solubility and Anticancer Potential of Mansonone G By β-Cyclodextrin-Based Host-Guest Complexation: A Computational and Experimental Study. Biomolecules 2019, 9, 545. [Google Scholar] [CrossRef] [PubMed]
  24. Matencio, A.; Dhakar, N.K.; Bessone, F.; Musso, G.; Cavalli, R.; Dianzani, C.; García-Carmona, F.; López-Nicolás, J.M.; Trotta, F. Study of oxyresveratrol complexes with insoluble cyclodextrin based nanosponges: Developing a novel way to obtain their complexation constants and application in an anticancer study. Carbohydr. Polym. 2020, 231, 115763. [Google Scholar] [CrossRef] [PubMed]
  25. Rezaei, A.; Varshosaz, J.; Fesharaki, M.; Farhang, A.; Jafari, S.M. Improving the solubility and in vitro cytotoxicity (anticancer activity) of ferulic acid by loading it into cyclodextrin nanosponges. Int. J. Nanomed. 2019, 14, 4589–4599. [Google Scholar] [CrossRef] [PubMed]
  26. Dhule, S.S.; Penfornis, P.; Frazier, T.; Walker, R.; Feldman, J.; Tan, G.; He, J.; Alb, A.; John, V.; Pochampally, R. Curcumin-loaded γ-cyclodextrin liposomal nanoparticles as delivery vehicles for osteosarcoma. Nanomed. Nanotechnol. Biol. Med. 2012, 8, 440–451. [Google Scholar] [CrossRef] [PubMed]
  27. Jyoti, K.; Bhatia, R.K.; Martis, E.A.F.; Coutinho, E.C.; Jain, U.K.; Chandra, R.; Madan, J. Soluble curcumin amalgamated chitosan microspheres augmented drug delivery and cytotoxicity in colon cancer cells: In vitro and in vivo study. Colloids Surf. B Biointerfaces 2016, 148, 674–683. [Google Scholar] [CrossRef]
  28. Al-Qubaisi, M.S.; Rasedee, A.; Flaifel, M.H.; Eid, E.E.M.; Hussein-Al-Ali, S.; Alhassan, F.H.; Salih, A.M.; Hussein, M.Z.; Zainal, Z.; Sani, D.; et al. Characterization of thymoquinone/hydroxypropyl-β-cyclodextrin inclusion complex: Application to anti-allergy properties. Eur. J. Pharm. Sci. 2019, 133, 167–182. [Google Scholar] [CrossRef]
  29. Rimbach, G.; Fischer, A.; Schloesser, A.; Jerz, G.; Ikuta, N.; Ishida, Y.; Matsuzawa, R.; Matsugo, S.; Huebbe, P.; Terao, K. Anti-inflammatory properties of Brazilian green propolis encapsulated in a γ-cyclodextrin complex in mice fed a western-type diet. Int. J. Mol. Sci. 2017, 18, 1141. [Google Scholar] [CrossRef]
  30. Ren, L.; Ma, X.-L.; Wang, H.-L.; Li, R.; Cui, J.-J.; Yan, P.-J.; Wang, Y.-N.; Yu, X.-Y.; Du, P.; Yu, H.-Y.; et al. Prebiotic-like cyclodextrin assisted silybin on NAFLD through restoring liver and gut homeostasis. J. Control. Release 2022, 348, 825–840. [Google Scholar] [CrossRef]
  31. da Silva, F.V.; de Barros Fernandes, H.; Oliveira, I.S.; Viana, A.F.S.C.; da Costa, D.S.; Lopes, M.T.P.; de Lira, K.L.; Quintans-Júnior, L.J.; de Sousa, A.A.; de Cássia Meneses Oliveira, R. Beta-cyclodextrin enhanced gastroprotective effect of (−)-linalool, a monoterpene present in rosewood essential oil, in gastric lesion models. Naunyn.-Schmiedeberg’s Arch. Pharmacol. 2016, 389, 1245–1251. [Google Scholar] [CrossRef]
  32. Matsuura, R.; Kawamura, A.; Matsumoto, Y.; Iida, Y.; Kanayama, M.; Kurokawa, M.; Aida, Y. Epigallocatechin Gallate Stabilized by Cyclodextrin Inactivates Influenza Virus and Human Coronavirus 229E. Microorganisms 2022, 10, 1796. [Google Scholar] [CrossRef] [PubMed]
  33. Silvestro, S.; Chiricosta, L.; Gugliandolo, A.; Iori, R.; Rollin, P.; Perenzoni, D.; Mattivi, F.; Bramanti, P.; Mazzon, E. The Moringin/α-CD Pretreatment Induces Neuroprotection in an In Vitro Model of Alzheimer’s Disease: A Transcriptomic Study. Curr. Issues Mol. Biol. 2021, 43, 197–214. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, Q.; Guan, J. Method for Improving Curcumin Stability. Patent No. CN110935033A, 10 December 2019. [Google Scholar]
  35. Codruţa, S.; Avram, S.; Borcan, F.; Trandafirescu, C.; Dehelean, A.; Danciu, C. Complexes of Triterpenic Acids with Hydrophilic Cyclodextrins, with Optimized Bio-Pharmaceutical Properties. Patent No. RO131432A2, 29 April 2015. [Google Scholar]
  36. Tomulewicz, M. Combination of Betulin and Cyclodextrin for Use in the Treatment of Alzheimer’s Disease. Patent No. EP3744331A1, 13 May 2020. [Google Scholar]
  37. Nianzeng, X.; Qinxin, Z.; Feiya, Y.; Dong, C.; Lingquan, M. Pharmaceutical Composition and Application Thereof in Preparation of Products for Treating Prostatitis. Patent No. CN111588716A, 11 November 2020. [Google Scholar]
  38. Mozuraityte, R.; Kristinova, V.; Rustad, T. Oxidation of Food Components. In Encyclopedia of Food and Health; Caballero, B., Finglas, P.M., Toldrá, F., Eds.; Academic Press: Oxford, UK, 2016; pp. 186–190. ISBN 978-0-12-384953-3. [Google Scholar]
  39. Ho, S.; Thoo, Y.Y.; Young, D.J.; Siow, L.F. Cyclodextrin encapsulated catechin: Effect of pH, relative humidity and various food models on antioxidant stability. LWT Food Sci. Technol. 2017, 85, 232–239. [Google Scholar] [CrossRef]
  40. Nguyen, T.A.; Liu, B.; Zhao, J.; Thomas, D.S.; Hook, J.M. An investigation into the supramolecular structure, solubility, stability and antioxidant activity of rutin/cyclodextrin inclusion complex. Food Chem. 2013, 136, 186–192. [Google Scholar] [CrossRef] [PubMed]
  41. Kfoury, M.; Auezova, L.; Greige-Gerges, H.; Fourmentin, S. Encapsulation in cyclodextrins to widen the applications of essential oils. Environ. Chem. Lett. 2019, 17, 129–143. [Google Scholar] [CrossRef]
  42. Zhu, G.; Zhu, G.; Xiao, Z. A review of the production of slow-release flavor by formation inclusion complex with cyclodextrins and their derivatives. J. Incl. Phenom. Macrocycl. Chem. 2019, 95, 17–33. [Google Scholar] [CrossRef]
  43. Sedaghat Doost, A.; Nikbakht Nasrabadi, M.; Kassozi, V.; Nakisozi, H.; Van der Meeren, P. Recent advances in food colloidal delivery systems for essential oils and their main components. Trends Food Sci. Technol. 2020, 99, 474–486. [Google Scholar] [CrossRef]
  44. Waleczek, K.J.; Marques, H.M.C.; Hempel, B.; Schmidt, P.C. Phase solubility studies of pure (−)-α-bisabolol and camomile essential oil with β-cyclodextrin. Eur. J. Pharm. Biopharm. 2003, 55, 247–251. [Google Scholar] [CrossRef]
  45. European Commission. Commission regulation (EU) No 231/2012 of 9 March 2012 laying down specifications for food additives listed in Annexes II and III to Regulation (EC) No 1333/2008 of the European Parliament and of the Council. In force. Off. J. Eur. Union 2012, L83, 1–295. [Google Scholar]
  46. European Parliament and the Council of the European Union. Regulation (EC) No 1333/2008 of the European Parliament and of the Council of 16 December 2008 on food additives. In force. Off. J. Eur. Union 2008, L354, 16–33. [Google Scholar]
  47. Jansook, P.; Kurkov, S.V.; Loftsson, T. Cyclodextrins as solubilizers: Formation of complex aggregates. J. Pharm. Sci. 2010, 99, 719–729. [Google Scholar] [CrossRef] [PubMed]
  48. Velázquez-Contreras, F.; Zamora-Ledezma, C.; López-González, I.; Meseguer-Olmo, L.; Núñez-Delicado, E.; Gabaldón, J.A. Cyclodextrins in Polymer-Based Active Food Packaging: A Fresh Look at Nontoxic, Biodegradable, and Sustainable Technology Trends. Polymers 2021, 14, 104. [Google Scholar] [CrossRef] [PubMed]
  49. Atarés, L.; Chiralt, A. Essential oils as additives in biodegradable films and coatings for active food packaging. Trends Food Sci. Technol. 2016, 48, 51–62. [Google Scholar] [CrossRef]
  50. Zhou, Z.; Liu, Y.; Liu, Z.; Fan, L.; Dong, T.; Jin, Y.; Saldaña, M.D.A.; Sun, W. Sustained-release antibacterial pads based on nonwovens polyethylene terephthalate modified by β-cyclodextrin embedded with cinnamaldehyde for cold fresh pork preservation. Food Packag. Shelf Life 2020, 26, 100554. [Google Scholar] [CrossRef]
  51. Cui, H.; Wang, Y.; Li, C.; Chen, X.; Lin, L. Antibacterial efficacy of Satureja montana L. essential oil encapsulated in methyl-β-cyclodextrin/soy soluble polysaccharide hydrogel and its assessment as meat preservative. LWT 2021, 152, 112427. [Google Scholar] [CrossRef]
  52. Yang, P.; Shi, Y.; Li, D.; Chen, R.; Zheng, M.; Ma, K.; Xu, N.; Dong, L.; Li, T. Antimicrobial and Mechanical Properties of β-Cyclodextrin Inclusion with Octyl Gallate in Chitosan Films and their Application in Fresh Vegetables. Food Biophys. 2022, 17, 598–611. [Google Scholar] [CrossRef]
  53. Chen, H.; Li, L.; Ma, Y.; Mcdonald, T.P.; Wang, Y. Development of active packaging film containing bioactive components encapsulated in β-cyclodextrin and its application. Food Hydrocoll. 2019, 90, 360–366. [Google Scholar] [CrossRef]
  54. Wu, J.; Li, J.; Xu, F.; Zhou, A.; Zeng, S.; Zheng, B.; Lin, S. Effective Preservation of Chilled Pork Using Photodynamic Antibacterial Film Based on Curcumin-β-Cyclodextrin Complex. Polymers 2023, 15, 1023. [Google Scholar] [CrossRef]
  55. Erceg, T.; Šovljanski, O.; Stupar, A.; Ugarković, J.; Aćimović, M.; Pezo, L.; Tomić, A.; Todosijević, M. A comprehensive approach to chitosan-gelatine edible coating with β-cyclodextrin/lemongrass essential oil inclusion complex—Characterization and food application. Int. J. Biol. Macromol. 2023, 228, 400–410. [Google Scholar] [CrossRef]
  56. Wu, H.; Ao, X.; Liu, J.; Zhu, J.; Bi, J.; Hou, H.; Hao, H.; Zhang, G. A Bioactive Chitosan-Based Film Enriched with Benzyl Isothiocyanate/α-Cyclodextrin Inclusion Complex and Its Application for Beef Preservation. Foods 2022, 11, 2687. [Google Scholar] [CrossRef]
  57. Lin, L.; Liao, X.; Li, C.; Abdel-Samie, M.A.; Siva, S.; Cui, H. Cold nitrogen plasma modified cuminaldehyde/β-cyclodextrin inclusion complex and its application in vegetable juices preservation. Food Res. Int. 2021, 141, 110132. [Google Scholar] [CrossRef]
  58. Amani, F.; Rezaei, A.; Kharazmi, M.S.; Jafari, S.M. Loading ferulic acid into β-cyclodextrin nanosponges; antibacterial activity, controlled release and application in pomegranate juice as a copigment agent. Colloids Surfaces A Physicochem. Eng. Asp. 2022, 649, 129454. [Google Scholar] [CrossRef]
  59. Wang, Y.; Du, Y.-T.; Xue, W.-Y.; Wang, L.; Li, R.; Jiang, Z.-T.; Tang, S.-H.; Tan, J. Enhanced preservation effects of clove (Syzygium aromaticum) essential oil on the processing of Chinese bacon (preserved meat products) by beta cyclodextrin metal organic frameworks (β-CD-MOFs). Meat Sci. 2023, 195, 108998. [Google Scholar] [CrossRef]
  60. Ghorbanzade, T.; Akhavan-Mahdavi, S.; Kharazmi, M.S.; Ibrahim, S.A.; Jafari, S.M. Loading of fish oil into β-cyclodextrin nanocomplexes for the production of a functional yogurt. Food Chem. X 2022, 15, 100406. [Google Scholar] [CrossRef] [PubMed]
  61. Silva, A.F.R.; Monteiro, M.; Resende, D.; Braga, S.S.; Coimbra, M.A.; Silva, A.M.S.; Cardoso, S.M. Inclusion Complex of Resveratrol with γ-Cyclodextrin as a Functional Ingredient for Lemon Juices. Foods 2020, 10, 16. [Google Scholar] [CrossRef]
  62. Zhong, Y.; Han, P.; Sun, S.; An, N.; Ren, X.; Lu, S.; Wang, Q.; Dong, J. Effects of apple polyphenols and hydroxypropyl-β-cyclodextrin inclusion complexes on the oxidation of myofibrillar proteins and microstructures in lamb during frozen storage. Food Chem. 2022, 375, 131874. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Z.; Guo, C.; Li, D.; Zhou, D.; Liu, D.; Zhu, B. Nanoprecipitates of γ-cyclodextrin/epigallocatechin-3-gallate inclusion complexes as efficient antioxidants for preservation of shrimp surimi products: Synthesis, performance and mechanism. J. Sci. Food Agric. 2023, 103, 3129–3138. [Google Scholar] [CrossRef]
  64. Pais, J.M.; Pereira, B.; Paz, F.A.A.; Cardoso, S.M.; Braga, S.S. Solid γ-Cyclodextrin Inclusion Compound with Gingerols, a Multi-Component Guest: Preparation, Properties and Application in Yogurt. Biomolecules 2020, 10, 344. [Google Scholar] [CrossRef] [PubMed]
  65. Sun, C.; Cao, J.; Wang, Y.; Chen, J.; Huang, L.; Zhang, H.; Wu, J.; Sun, C. Ultrasound-mediated molecular self-assemble of thymol with 2-hydroxypropyl-β-cyclodextrin for fruit preservation. Food Chem. 2021, 363, 130327. [Google Scholar] [CrossRef]
  66. He, J.; Guo, F.; Lin, L.; Chen, H.; Chen, J.; Cheng, Y.; Zheng, Z.-P. Investigating the oxyresveratrol β-cyclodextrin and 2-hydroxypropyl-β-cyclodextrin complexes: The effects on oxyresveratrol solution, stability, and antibrowning ability on fresh grape juice. LWT 2019, 100, 263–270. [Google Scholar] [CrossRef]
  67. Garcia-Sotelo, D.; Silva-Espinoza, B.; Perez-Tello, M.; Olivas, I.; Alvarez-Parrilla, E.; González-Aguilar, G.A.; Ayala-Zavala, J.F. Antimicrobial activity and thermal stability of rosemary essential oil:β−cyclodextrin capsules applied in tomato juice. LWT 2019, 111, 837–845. [Google Scholar] [CrossRef]
  68. Upreti, M.; Prakash, I.; Chen, Y.L. Solubility Enhanced Terpene Glycoside(s). Patent No. US20110195161, 8 February 2011. [Google Scholar]
  69. Smit, R. Extension of Beverage Shelf-Stability by Solute-Ligand Complexes. Patent No. EP2312958, 27 July 2009. [Google Scholar]
  70. Bogkhani, N.; Gebreselassi, P. Encapsulated Compositions and Methods of Preparation. Patent No. US20060068059, 30 September 2004. [Google Scholar]
  71. Bogkhani, N.; Gebreselassi, P. Encapsulated Compositions and Its Production Method. Patent No. RU2366292C2, 30 September 2005. [Google Scholar]
  72. Gebreselassi, P.; Luo Shiukh, D.; Bogkhani, N. Stain Removing Chewing Gum Composition. Patent No. US20050008732, 11 July 2003. [Google Scholar]
  73. Snabe, T.; Thompson, B.W.; Mallory, S.L. Natamycin-Cyclodextrin Complexes for Use in Foodstuff, Process for Their Manufacture and Use Thereof. Patent No. US20120196003, 28 January 2011. [Google Scholar]
  74. Mortensen, B.; Jansson, S.T.K. Complexes of Cyclodextrins and Carotenoids. Patent No. AU2003258890, 4 July 2003. [Google Scholar]
  75. Wüpper, S.; Lüersen, K.; Rimbach, G. Cyclodextrins, Natural Compounds, and Plant Bioactives-A Nutritional Perspective. Biomolecules 2021, 11, 401. [Google Scholar] [CrossRef]
  76. Prosek, M.; Smidovnik, A.; Fir, M.; Strazisar, M.; Golc Wondra, A.; Andrensek, S.; Zmitek, J. Water-Soluble Coenzyme Q10 Inclusion Complex with Beta-Cyclodextrin, Process of Preparing, and Use Thereof. Patent No. EP1755683, 10 May 2005. [Google Scholar]
  77. Bartlett, M.; Mastaloudis, A.; Smitdt, C.; Poole, S.J. Nanosized Carotenoid Cyclodextrin Complexes. Patent No. US20070191307, 4 October 2006. [Google Scholar]
  78. Arigony Souto, A. Process for the Preparation of a Water-Soluble Complex Having Resveratrol Compounds; Products Comprising Said Complex; and Uses Thereof. Patent No. US20100204179, 23 July 2008. [Google Scholar]
  79. Klaveness, J.; Brudeli, B.; Rongved, P. Cyclodextrins for Administering Fatty Acids in Tablets. Patent No. EP2164520, 2 June 2008. [Google Scholar]
  80. Nowak, J.K.; Sobkowiak, P.; Drzymała-Czyż, S.; Krzyżanowska-Jankowska, P.; Sapiejka, E.; Skorupa, W.; Pogorzelski, A.; Nowicka, A.; Wojsyk-Banaszak, I.; Kurek, S.; et al. Fat-Soluble Vitamin Supplementation Using Liposomes, Cyclodextrins, or Medium-Chain Triglycerides in Cystic Fibrosis: A Randomized Controlled Trial. Nutrients 2021, 13, 4554. [Google Scholar] [CrossRef] [PubMed]
  81. Gaur, S.; Lopez, E.C.; Ojha, A.; Andrade, J.E. Functionalization of Lipid-Based Nutrient Supplement with β-Cyclodextrin Inclusions of Oregano Essential Oil. J. Food Sci. 2018, 83, 1748–1756. [Google Scholar] [CrossRef] [PubMed]
  82. Stasiłowicz, A.; Tykarska, E.; Lewandowska, K.; Kozak, M.; Miklaszewski, A.; Kobus-Cisowska, J.; Szymanowska, D.; Plech, T.; Jenczyk, J.; Cielecka-Piontek, J. Hydroxypropyl-β-cyclodextrin as an effective carrier of curcumin—Piperine nutraceutical system with improved enzyme inhibition properties. J. Enzyme Inhib. Med. Chem. 2020, 35, 1811–1821. [Google Scholar] [CrossRef]
  83. Okawara, M.; Tokudome, Y.; Todo, H.; Sugibayashi, K.; Hashimoto, F. Enhancement of diosgenin distribution in the skin by cyclodextrin complexation following oral administration. Biol. Pharm. Bull. 2013, 36, 36–40. [Google Scholar] [CrossRef]
  84. Ferreira, L.; Mascarenhas-Melo, F.; Rabaça, S.; Mathur, A.; Sharma, A.; Giram, P.S.; Pawar, K.D.; Rahdar, A.; Raza, F.; Veiga, F.; et al. Cyclodextrin-based dermatological formulations: Dermopharmaceutical and cosmetic applications. Colloids Surf. B Biointerfaces 2023, 221, 113012. [Google Scholar] [CrossRef]
  85. Hans-Jurgen, B.; Eckhard, S. Applications of cyclodextrins in cosmetic products: A review. J. Cosmet. Sci. 2002, 53, 185–192. [Google Scholar]
  86. Santos, A.C.; Morais, F.; Simões, A.; Pereira, I.; Sequeira, J.A.D.; Pereira-Silva, M.; Veiga, F.; Ribeiro, A. Nanotechnology for the development of new cosmetic formulations. Expert Opin. Drug Deliv. 2019, 16, 313–330. [Google Scholar] [CrossRef]
  87. Crini, G.; Fenyvesi, É.; Szente, L. Outstanding contribution of Professor József Szejtli to cyclodextrin applications in foods, cosmetics, drugs, chromatography and biotechnology: A review. Environ. Chem. Lett. 2021, 19, 2619–2641. [Google Scholar] [CrossRef]
  88. Pereira, A.G.; Carpena, M.; Oliveira, P.G.; Mejuto, J.C.; Prieto, M.A.; Gandara, J.S. Main applications of cyclodextrins in the food industry as the compounds of choice to form host–guest complexes. Int. J. Mol. Sci. 2021, 22, 1339. [Google Scholar] [CrossRef] [PubMed]
  89. Singh, A.; Kaler, A.; Singh, V.; Patil, R.; Banerjee, U.C. Cyclodextrins and Biotechnological Applications. In Cyclodextrins in Pharmaceutics, Cosmetics, and Biomedicine: Current and Future Industrial Applications; John Wiley & Sons: Hoboken, NJ, USA, 2011; pp. 275–285. [Google Scholar]
  90. Braga, S.S.; Pais, J. Getting under the skin: Cyclodextrin inclusion for the controlled delivery of active substances to the dermis. In Design of Nanostructures for Versatile Therapeutic Applications, 1st ed.; Grumezescu, A.M., Ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2018; pp. 407–449. [Google Scholar]
  91. Suvarna, V.; Bore, B.; Bhawar, C.; Mallya, R. Complexation of phytochemicals with cyclodextrins and their derivatives—An update. Biomed. Pharmacother. 2022, 149, 112862. [Google Scholar] [CrossRef] [PubMed]
  92. Numanoǧlu, U.; Şen, T.; Tarimci, N.; Kartal, M.; Koo, O.M.Y.; Önyüksel, H. Use of cyclodextrins as a cosmetic delivery system for fragrance materials: Linalool and benzyl acetate. AAPS PharmSciTech 2007, 8, 34–42. [Google Scholar] [CrossRef] [PubMed]
  93. Aytac, Z.; Yildiz, Z.I.; Kayaci-Senirmak, F.; Tekinay, T.; Uyar, T. Electrospinning of cyclodextrin/linalool-inclusion complex nanofibers: Fast-dissolving nanofibrous web with prolonged release and antibacterial activity. Food Chem. 2017, 231, 192–201. [Google Scholar] [CrossRef]
  94. Xiao, Z.; Zhang, Y.; Niu, Y.; Ke, Q.; Kou, X. Cyclodextrins as carriers for volatile aroma compounds: A review. Carbohydr. Polym. 2021, 269, 118292. [Google Scholar] [CrossRef]
  95. Arana-Sánchez, A.; Estarrón-Espinosa, M.; Obledo-Vázquez, E.N.; Padilla-Camberos, E.; Silva-Vázquez, R.; Lugo-Cervantes, E. Antimicrobial and antioxidant activities of Mexican oregano essential oils (Lippia graveolens H. B. K.) with different composition when microencapsulated inβ-cyclodextrin. Lett. Appl. Microbiol. 2010, 50, 585–590. [Google Scholar] [CrossRef]
  96. Ruksiriwanich, W.; Sirithunyalug, J.; Khantham, C.; Leksomboon, K.; Jantrawut, P. Skin penetration and stability enhancement of Celastrus paniculatus seed oil by 2-hydroxypropyl-β-cyclodextrin inclusion complex for cosmeceutical applications. Sci. Pharm. 2018, 86, 33. [Google Scholar] [CrossRef]
  97. Marijan, M.; Tomić, D.; Strawa, J.W.; Jakupović, L.; Inić, S.; Jug, M.; Tomczyk, M.; Zovko Končić, M. Optimization of Cyclodextrin-Assisted Extraction of Phenolics from Helichrysum italicum for Preparation of Extracts with Anti-Elastase and Anti-Collagenase Properties. Metabolites 2023, 13, 257. [Google Scholar] [CrossRef]
  98. Wang, W.; Yang, Y.; Tang, K. Selective Extraction of Glabridin from Glycyrrhiza Glabra Crude Extract by sulfobutylether -β-CD in a Ternary Solvent System. Process Biochem. 2023, 129, 1–10. [Google Scholar] [CrossRef]
  99. Kalouta, K.; Eleni, P.; Boukouvalas, C.; Vassilatou, K.; Krokida, M. Dynamic mechanical analysis of novel cosmeceutical facial creams containing nano-encapsulated natural plant and fruit extracts. J. Cosmet. Dermatol. 2020, 19, 1146–1154. [Google Scholar] [CrossRef]
  100. Spanidi, E.; Athanasopoulou, S.; Liakopoulou, A.; Chaidou, A.; Hatziantoniou, S.; Gardikis, K. Royal Jelly Components Encapsulation in a Controlled Release System—Skin Functionality, and Biochemical Activity for Skin Applications. Pharmaceuticals 2022, 15, 907. [Google Scholar] [CrossRef]
  101. Dias, P.H.; Scopel, M.; Martiny, S.; Bianchi, S.E.; Bassani, V.L.; Zuanazzi, J.A.S. Hydroxypropyl-β-cyclodextrin-containing hydrogel enhances skin formononetin permeation/retention. J. Pharm. Pharmacol. 2018, 70, 865–873. [Google Scholar] [CrossRef] [PubMed]
  102. Spanidi, E.; Karapetsas, A.; Voulgaridou, G.P.; Letsiou, S.; Aligiannis, N.; Tsochantaridis, I.; Kynigopoulos, S.; Lambropoulou, M.; Mourtzinos, I.; Pappa, A.; et al. A new controlled release system for propolis polyphenols and its biochemical activity for skin applications. Plants 2021, 10, 420. [Google Scholar] [CrossRef] [PubMed]
  103. Dalla, E.; Koumentakou, I.; Bikiaris, N.; Balla, E.; Lykidou, S.; Nikolaidis, N. Formulation, Characterization and Evaluation of Innovative O/W Emulsions Containing Curcumin Derivatives with Enhanced Antioxidant Properties. Antioxidants 2022, 11, 2271. [Google Scholar] [CrossRef] [PubMed]
  104. Wu, L.; Liu, G.; Wang, W.; Liu, R.; Liao, L.; Cheng, N.; Li, W.; Zhang, W.; Ding, D. Cyclodextrin-modified CeO2 nanoparticles as a multifunctional nanozyme for combinational therapy of psoriasis. Int. J. Nanomed. 2020, 15, 2515–2527. [Google Scholar] [CrossRef]
  105. Kumar, S.; Singh, K.K.; Rao, R. Enhanced anti-psoriatic efficacy and regulation of oxidative stress of a novel topical babchi oil (Psoralea corylifolia) cyclodextrin-based nanogel in a mouse tail model. J. Microencapsul. 2019, 36, 140–155. [Google Scholar] [CrossRef]
  106. Wathoni, N.; Motoyama, K.; Higashi, T.; Okajima, M.; Kaneko, T.; Arima, H. Enhancement of curcumin wound healing ability by complexation with 2-hydroxypropyl-γ-cyclodextrin in sacran hydrogel film. Int. J. Biol. Macromol. 2017, 98, 268–276. [Google Scholar] [CrossRef]
  107. Pipkin, J.D.; Rajewski, R.; Mainous, B. Compositions Containing Silymarin and Sulfoalkyl Ether Cyclodextrin and Methods of Using the Same. Patent No. EP3270941, 18 March 2016. [Google Scholar]
  108. Deng, Y.; Pan, Z.; Bernard, A.-L.S.; Wu, S. Aqueous Compositions with Mangiferin for Cosmetic Applications. Patent No. US20190159989, 30 November 2011. [Google Scholar]
  109. Mourtzinos, I.; Koutsianas, N.; Dragani, P.; Patera, A. Extraction And Formation of Inclusion Complexes of Propolis Active Components with Hydroxypropyl—Beta—Cyclodextrin. Patent No. EP2646041, 30 November 2011. [Google Scholar]
  110. Gardikis, K.; Koutsianas, N.; Patera, A.; Dragani, P.; Tsoukalas, A.-I.; Letsiou, S. Method for Preparing a Stable Controlled Release Propolis Colloidal Dispersion System for Various Uses. Patent No. EP3380081, 23 November 2015. [Google Scholar]
  111. Gardikis, K. Preparation Method of a Colloidal System of Stabilisation and Contolled Release of Royal Jelly Components for Various Uses. Patent No. EP4117693, 29 October 2020. [Google Scholar]
  112. Loftsson, T.; Duchêne, D. Cyclodextrins and their pharmaceutical applications. Int. J. Pharm. 2007, 329, 1–11. [Google Scholar] [CrossRef]
  113. Bonini, M.; Rossi, S.; Karlsson, G.; Almgren, M.; Nostro, P.L.; Baglioni, P. Self-assembly of β-cyclodextrin in water. Part 1: Cryo-TEM and dynamic and static light scattering. Langmuir 2006, 22, 1478–1484. [Google Scholar] [CrossRef]
  114. Okamoto, H.; Komatsu, H.; Hashida, M.; Sezaki, H. Effects of β-cyclodextrin and di-O-methyl-β-cyclodextrin on the percutaneous absorption of butylparaben, indomethacin and sulfanilic acid. Int. J. Pharm. 1986, 30, 35–45. [Google Scholar] [CrossRef]
  115. Loftsson, T.; Vogensen, S.B.; Brewster, M.E.; Konrádsdóttir, F. Effects of cyclodextrins on drug delivery through biological membranes. J. Pharm. Sci. 2007, 96, 2532–2546. [Google Scholar] [CrossRef] [PubMed]
  116. Andreia, A.; Helena, R.; Sandra, S. Carrier-Mediated Dermal Delivery: Applications in the Prevention and Treatment of Skin Disorders, 1st ed.; Jenny Stanford Publishing: Singapore, 2017; ISBN 9789814745581. [Google Scholar]
  117. Piel, G.; Moutard, S.; Uhoda, E.; Pilard, F.; Piérard, G.E.; Perly, B.; Delattre, L.; Evrard, B. Skin compatibility of cyclodextrins and their derivatives: A comparative assessment using a corneoxenometry bioassay. Eur. J. Pharm. Biopharm. 2004, 57, 479–482. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Trends in the number of publications that contain the words “cyclodextrins” and “delivery systems”; “cyclodextrins” and “delivery systems” and “medicine”; “cyclodextrins” and “delivery systems” and “food”; and “cyclodextrins” and “delivery systems” and “cosmetics” in the title, abstract or keyword fields, in the period 2000–2023 (Science Direct search carried out on 28 August 2023).
Figure 1. Trends in the number of publications that contain the words “cyclodextrins” and “delivery systems”; “cyclodextrins” and “delivery systems” and “medicine”; “cyclodextrins” and “delivery systems” and “food”; and “cyclodextrins” and “delivery systems” and “cosmetics” in the title, abstract or keyword fields, in the period 2000–2023 (Science Direct search carried out on 28 August 2023).
Pharmaceuticals 16 01274 g001
Table 5. Applications of CD-based ICs in cosmetics and their main effects.
Table 5. Applications of CD-based ICs in cosmetics and their main effects.
CD TypeBioactive Compound/Guest MoietySystem/ModelEffects/Key FindingsReference
β-CDs and HPβCDLinaloolGel formulationsIncreased the water solubility, improved handling of raw materials (liquid fragrance material to be turned into powder). Controlled release of bioactive compounds and their stability within the formula.[92]
HPβCDCelastrus paniculatus seed oil (CPSO)Serum and gel basePhysical stability of formulations containing CPSO with HPβCD after 3 months of storage, with the percentage of oleic acid maintained above 80% of the initial amount. Higher skin penetration of oleic acid is shown compared to other formulations. The formula exhibits appropriate viscosities for use in cosmetic products.[96]
β-CDsTea tree oilCosmeceutical facial creamsStable formulas mainly in rheological properties[99]
HPβCDRoyal jellyCosmeceutical facial creamsEncapsulates the sensitive components of royal jelly (10-HDA), eliminating its stability disadvantages while at the same time allowing time-controlled release which could prove useful for skin applications as indicated by the in-cell experiments[100]
HPβCD and MβCDIsoflavones (formononetin and biochanin A)Hydrogel formulationsSignificant increase in the penetration of isoflavones (formononetin and biochanin A) into the epidermis and dermis with the use of HPβCD in hydrogels, with potential use in cosmetic formulations for the prevention of skin aging[101]
HPβCDPropolisCosmeceutical facial creamsEncapsulation of propolis polyphenols in a physicochemically stable system with a controlled release rate. It maintains the antioxidant, anti-mutagenic and anti-aging properties of propolis polyphenols at levels similar to a methanolic extract[102]
β-CDCurcuminSemisolid Oil in Water (O/W) emulsionsFormulas containing curcumin and β-CD showed greater antioxidant capacity and improved viscosity as well as stability, but presented a low rate of antimicrobial activity[103]
β-CDsCeria NPs (CeNPs), natural antioxidant enzymes Increase biocompatibility, water solubility and antioxidant capacity, anti-psoriatic effects.[104]
β-CDBabchi Oil—BONanostructure gelReduce oxidative stress, anti-psoriatic effects[105]
HPγCDCurcuminHydrogel filmWound healing[106]
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

Christaki, S.; Spanidi, E.; Panagiotidou, E.; Athanasopoulou, S.; Kyriakoudi, A.; Mourtzinos, I.; Gardikis, K. Cyclodextrins for the Delivery of Bioactive Compounds from Natural Sources: Medicinal, Food and Cosmetics Applications. Pharmaceuticals 2023, 16, 1274. https://doi.org/10.3390/ph16091274

AMA Style

Christaki S, Spanidi E, Panagiotidou E, Athanasopoulou S, Kyriakoudi A, Mourtzinos I, Gardikis K. Cyclodextrins for the Delivery of Bioactive Compounds from Natural Sources: Medicinal, Food and Cosmetics Applications. Pharmaceuticals. 2023; 16(9):1274. https://doi.org/10.3390/ph16091274

Chicago/Turabian Style

Christaki, Stamatia, Eleni Spanidi, Eleni Panagiotidou, Sophia Athanasopoulou, Anastasia Kyriakoudi, Ioannis Mourtzinos, and Konstantinos Gardikis. 2023. "Cyclodextrins for the Delivery of Bioactive Compounds from Natural Sources: Medicinal, Food and Cosmetics Applications" Pharmaceuticals 16, no. 9: 1274. https://doi.org/10.3390/ph16091274

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