Next Article in Journal
Boosting the In Vivo Transdermal Bioavailability of Asenapine Maleate Using Novel Lavender Oil-Based Lipid Nanocapsules for Management of Schizophrenia
Previous Article in Journal
In Vitro Studies of Pegylated Magnetite Nanoparticles in a Cellular Model of Viral Oncogenesis: Initial Studies to Evaluate Their Potential as a Future Theranostic Tool
Previous Article in Special Issue
Metformin-NSAIDs Molecular Salts: A Path towards Enhanced Oral Bioavailability and Stability
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Formulation Approaches to Crystalline Status Modification for Carotenoids: Impacts on Dissolution, Stability, Bioavailability, and Bioactivities

1
School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
2
Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
3
Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
4
Drug Development and Value Creation Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
5
Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 16 December 2022 / Revised: 30 January 2023 / Accepted: 30 January 2023 / Published: 1 February 2023
(This article belongs to the Special Issue Applications of Crystal Engineering in Drug Delivery)

Abstract

:
Carotenoids, including carotenes and xanthophylls, have been identified as bioactive ingredients in foods and are considered to possess health-promoting effects. From a biopharmaceutical perspective, several physicochemical characteristics, such as scanty water solubility, restricted dissolution, and susceptibility to oxidation may influence their oral bioavailability and eventually, their effectiveness. In this review, we have summarized various formulation approaches that deal with the modification of crystalline status for carotenoids, which may improve their physicochemical properties, oral absorption, and biological effects. The mechanisms involving crystalline alteration and the typical methods for examining crystalline states in the pharmaceutical field have been included, and representative formulation approaches are introduced to unriddle the mechanisms and effects more clearly.

Graphical Abstract

1. Introduction

Carotenoids are composed of compounds containing typically 40 carbon atoms and are synthesized by plants or microorganisms. The structure of carotenoids commonly includes a central carbon chain with several conjugated double bonds and they partially have different cyclic or acyclic end groups [1]. Carotenoids are divided into carotenes and xanthophylls depending on the chemical structure as presented in Figure 1. Carotenes, such as α-carotene, β-carotene, and lycopene, only contain carbon and hydrogen in their structure without oxygen atoms. Xanthophylls, such as astaxanthin, β-cryptoxanthin, lutein, and zeaxanthin, are the other type; they are carotenoids containing one or more oxygen atoms in their structure. The bioactivities of carotenoids have been demonstrated to be associated with chemical structures, such as the number of conjugated double bonds and the types of functional groups at the ends [2].
Carotenoids exhibit well-known anti-oxidative activities and are most likely involved in scavenging the singlet oxygen and peroxy radicals [3]. Reactive oxygen species (ROS) are generated during normal metabolism and engaged in enzymatic reactions, mitochondrial electron transport and signal transduction. Excessive ROS would damage biologically essential factors and elevate the risk of degenerative diseases [4]. Therefore, carotenoids are considered excellent antioxidants that benefit various diseases associated with oxidative stress. The bioactivities of carotenoids have been reported in previous studies and are listed in Table 1.
Carotenoids commonly exist in crystalline states and exhibit an ordered intermolecular arrangement in the solid state. Various intermolecular interactions, such as hydrogen bonding and π-π stacking interactions, maintain the structure integrity and crystalline status of carotenoids [5,6]. The crystalline status of carotenoids contributes to the scant solubility and dissolution, which further restricts their oral bioavailability and health-promoting effects. Therefore, improving the solubility of carotenoids is a critical issue for achieving the desired plasma concentration in systemic circulation to attain the expected biological effects [7]. The higher lattice energy of crystalline compounds usually correlates with their poor solubilities, because the energy has to be overcome before the compound can be dissolved in the medium [8]. Carotenoids have poor aqueous solubility owing to the higher lattice energy. Thus, the crystalline status is a crucial factor for the application of carotenoids.
Table 1. The bioactivities of carotenoids that have been reported in previous studies.
Table 1. The bioactivities of carotenoids that have been reported in previous studies.
CarotenoidsDoseModelBioactivitiesReference
Astaxanthin1.0 mg/mouse/dayDiabetic C57BL/KsJ-db/db miceAnti-diabetic
(Blood glucose↓ and preservation of -cell function)
[9]
Astaxanthin2 mgHealthy womenImmune response improvement
(Mitogen-induced lymphoproliferation↑
Natural killer cell, total T and B cell↑
DNA damage biomarker↓)
[10]
Astaxanthin5 μMPrimary hippocampal neuronsTreatment of Hcy-mediated neurological disorders
(ROS and superoxide anion↓)
[11]
β-Carotene45 mg/dayHealthy older adultsImmunostimulant
(Total T cells and NK cell↑)
[12]
β-Carotene200 mg/KgMale albino miceAnticonvulsant activity
(Duration of general tonic–clonic seizures↓
General tonic–clonic seizures latency↑)
[13]
β-Carotene30 μMHuman prostate cancer cell line (PC-3 cell)Anticancer (cell viability: 51.4%)[14]
β-Carotene2.05 mg/KgMale albino miceTreatment of Alzheimer’s disease
(Acetylcholinesterase and amyloid β-protein↓)
[15]
β-Cryptoxanthin0.8 mg/Kg/dayMale miceAnti-obesity
(Adipocyte hypertrophy↓)
[16]
Fucoxanthin5 μMHuman fibroblastProtection against UVB radiation-induced oxidative stress
(ROS↓)
[17]
Fucoxanthin1.06-2.22%C57BL/6J miceAnti-obesity and anti-diabetic effects
(Body weight and white adipose tissue↓
MCP-1 expression↓ and Adrb3 and GLUT4↑)
[18]
Fucoxanthin0.2%C57BL/6N miceAnti-obesity
(Fatty acid β-oxidation activity and
lipogenic enzyme activities ↓)
[19]
Lutein and
Zeaxanthin
Oral:
lutein 100 ppm
zeaxanthin 6 ppm
Topical:
lutein 10 ppm
zeaxanthin 0.6 ppm
Healthy womenPhotoprotective
(Lipid peroxidation↓
skin lipid, skin hydration and skin elasticity↑)
[20]
Lutein and
Zeaxanthin
Lutein: 5%
zeaxanthin: 0.2%
β5−/− micePrevention of age-related retinal pigment epithelium actin damage
(4-hydroxynonenal-adduct formation, age-related cone and rod photoreceptor dysfunction ↓)
[21]
Lutein and
Zeaxanthin
Lutein:10 mg
Zeaxanthin: 2 mg
Healthy older adultsImprovement of cognitive function
(Macular pigment optical density, complex attention and cognitive flexibility domains↑)
[22]
Lycopene5 μg/mLFungal cell
(Candida albicans)
Antifungi
(Destruction of fungi membrane and inhibition of the normal budding process)
[23]
Lycopene2 μMRat cortical neuronsTreatment of Alzheimer’s disease
(Intracellular ROS and superoxide production↓)
[24]
Lycopene0.2 or 0.5 μMNeuronal SH-SY5Y cellsNeuron protection
(ROS↓ and mitochondrial dysfunction ↓)
[25]
Lycopene0.03% (w/w, mixed into normal chow)Male C57BL/6J miceTreatment of Alzheimer’s disease
(Memory loss behavior, amyloid plaques, amyloid precursor protein, neuronal β-secretase BACE1, inflammatory mediators and oxidative stress↓, α-secretase ADAM10↑)
[26]
Lycopene2 μMMice cerebral cortical neuronsNeuron protection
(Nerve growth factor, brain-derived neurotrophic factor, and vascular endothelial growth factor excretion↑ and anti-apoptosis)
[27]
Lycopene100 mg/KgFemale Sprague-Dawley ratsTreatment of vascular dementia
(Oxidative stress in hippocampus↓)
[28]
↑: enhancement. ↓: reduction. ROS: reactive oxygen species. Adrb3: β3-adrenergic receptor. GLUT4: glucose transporter 4.

2. Effects of Crystalline Status Modification on the Physicochemical Properties of Carotenoids

The chemical structures of carotenoids possess many chiral centers, which result in a variety of conformations. The cis-form (Z-form) and trans-form (E-form) have been demonstrated to affect the crystalline state and further have an impact on the physicochemical properties. In general, the all-trans carotenoid isomers are the most stable ones owing to their different Gibbs free energies and exist commonly in nature. Only 5-cis-lycopene was found to be more stable than all-trans-isomers [29]. The cis-form isomers display diverse properties compared to the trans-form isomers, such as a shallower color caused by the shorter maximum absorption wavelength and smaller extinction coefficient [30], reduction in the crystalline ratio, lower melting point, and poor stability [31]. Taking β-carotene as an example, Figure 2 shows the isomers of β-carotene and the maximum absorption wavelength of these compounds [32]. A previous study reported the transformation method and different properties of the E-form and Z-form carotenoids. The Z-form lycopene has been discovered to have a 4000-fold higher solubility compared to the E-form in ethanol [6]. The lower degree of crystallinity leads to higher solubility in bile acid micelles, and the higher solubility would further result in greater bioaccessibility. Interestingly, the ideal bioaccessibility cannot completely correlate with bioavailability [6]. The cellular uptake efficiency is a critical factor that influences bioavailability, and the efficiency depends on the molecular structure and hydrophobic properties. In previous studies, Yang et al. [33,34] found that the trans-lutein had better passive diffusion into enterocytes due to the linear structure, and the affinity to transporters in the intestine changed the cellular uptake efficiency as well because the relatively higher solubility of 9Z-astaxanthin caused poorer affinity to the transporter compared with 13Z-astaxanthin and the trans-isomer. This theory can also be applied to β-carotene. E-β-carotene was also reported to exhibit higher absorption than the Z-form in both in vitro [35] and in vivo studies [36]. Therefore, modification of the crystalline conformation impacts the solubility, dissolution, intestinal absorption, and further bioavailability of carotenoids, together with the bioactivity.
Micronized crystalline lutein has been prepared to improve its dissolution and oral bioavailability [37]. A wet-jet milling method with high mechanical force was applied to reduce the particle size, and the procedures also converted the crystalline form into the polymorphic state, which belonged to a metastable situation with higher energy. The effects of size reduction and crystalline transformation are beneficial to dissolution and oral absorption. Though the solubility may have effects on the transporter affinity, the absorption still requires the transition from solid form to solution [38]. In order to improve the aqueous solubility of active crystalline compounds, the alternation of the crystalline condition is the simplest method. Crystalline states can be classified into three types—polymorphic state, pseudo-polymorphic state, and amorphous state—the difference among the three states is shown in Figure 3. Polymorphs are the same constituents with different crystalline arrangements, and they often benefit solubility. Notably, the improvement is small due to the small energy difference between the polymorphs [39]. Pseudo-polymorphs include hydrates or solvates, and they are usually unwanted crystalline forms. In the case of solvates, this depends on the characteristics of solvents. On the other hand, water forms hydrogen bonds with the active compounds in hydrates, and the lattice enthalpy would escalate to decrease the solubility, accompanied by a lower bioavailability [5]. The amorphous state lacks the ordered arrangement of molecules, causing amorphs to have poor thermodynamic stability, and it is usually the first choice to greatly enhance the aqueous solubility [40]. The amorphous state is divided into molecularly pure, which only transfers the crystalline state of the active compounds and formulation. Both of them may increase the solubility; however, the stability and scale-up of the molecularly pure type are hard to achieve [41]. Therefore, preparing a formulation to transfer the crystal to another state is quite an important technique to enhance the solubility and oral absorption, and then exhibit the expected bioactivities. To distinguish the crystalline status in the formulation, detection methods have been introduced.

3. Methods for Examining the Crystalline Status

Powder X-ray diffraction (PXRD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), transmission electron microscopy (TEM), and scanning electron microscopy (SEM) are fundamental methods to determine the characterization of the crystal in pharmaceutical formulations; usually, more than two methods are adopted to reveal more reliable results.

3.1. PXRD

PXRD is a well-known method to qualify the crystalline state and quantify the crystallinity as well as crystal size in a formulation. Fingerprint data of APIs can be obtained by PXRD, and possible polymorphs can be identified. The intensities of diffraction peaks are positively related to crystallinity, and we can use them to calculate the percentage of amorphization. However, nanocrystals and amorphism are hard to differentiate because the low crystallinity leads to broad peaks (i.e., Scherrer broadening) [42]. The nanocrystal size can be calculated using the Scherrer equation (Equation (1)), and the formula is only viable for nanocrystals around 100–200 nm [43].
τ = kλ/(β cosθ)
where τ is the average size of the crystal; k is the shape factor; λ is the wavelength of the X-ray; β is the line broadening at half the maximum intensity in radians; θ is the Bragg angle.
Figure 4 shows the PXRD profiles of β-carotene and nanoformulations, where the API group presents sharp and strong peaks compared with the nanoformulation groups, indicating the API has been encapsulated in the formulations in an amorphous state.

3.2. Electron Microscopy—TEM and SEM

For nanoscale crystals, electron microscopy is applied to visualize the lattice. The crystal size and the structure of the lattice are provided by TEM. The sizes calculated using TEM may be larger than those from the Scherrer equation as particles seen in the TEM images are possibly not crystals, and the crystalline imperfections broaden the peaks in PXRD, causing calculation bias [44]. SEM is a technology suitable for morphology observation, and the relationship between phases such as erosion can be further acquired compared with TEM [45]. However, the two technologies are limited by the samples “seen” by the microscope, and only unilateral information may be obtained. Thus, more significant sample sizes and observed angles are needed to avoid sampling errors and to obtain the whole morphology.

3.3. Thermal Methods—DSC and TGA

Thermal methods detect the thermal behavior of the whole sample, and they will not encounter issues with the difference between the surface and the core or the sampling bias [46]. DSC measures the heat required for the temperature increase in the samples; the heat absorption from melt formation or release due to the crystallization will be detected [42], and Tm (melting point), Tg (glass transition temperature), Tc (crystallization temperature), and Td (degradation temperature) can be obtained [47]. The endothermic Tm peak disappears once the crystals convert into amorphs, and the concept can be applied to confirm the encapsulation of amorphous APIs in formulations [48]. Taking DSC thermograms (Figure 5) as an example, the peak standing for the melting point of β-carotene at 186 °C was observed in the crystalline API and physical mixture, and the peak disappeared in the formulation group. However, it is hard to distinguish the amorph merely from the thermogram when the degree of crystallization is relatively high, and a simple formula (Equation (2)) can be used to calculate the amorphous content [49].
Amorphous   content = Δ H f amorphous Δ H f crystal
where Δ H f amorphous represents the enthalpy of the fusion of the amorphous fraction, and Δ H f crystal represents the enthalpy of the fusion of the crystalline fraction.
TGA measures the weight loss resulting from heating, and this technology can be used to investigate crystals containing volatile substances [5]. It is also often conducted combined with DSC to further confirm the thermal behavior. The crystal and amorph can be distinguished by the different weight loss under the same temperature [50], which can be used to prove the complex formation [51].

4. Effects of Preparation Factors on Crystalline Status of Active Pharmaceutical Ingredients

Turning active pharmaceutical ingredients (APIs) into amorphous or polymorph states is expected to change their biopharmaceutical properties, including their dissolution rate and bioavailability, which can be accomplished using various manufacturing processes. Major preparation factors that affect the crystalline status are summarized in Figure 6.

4.1. Excipients

Crystallization often includes two major steps, namely, nucleation and growth; the addition of excipients can manipulate the crystallization of APIs. The rate of nucleation is usually positively related to molecular mobility, suggesting that the restriction of molecular mobility through intermolecular interaction between excipients and APIs can also affect the crystallization of APIs.
Poly(ethylene oxide) as a plasticizer increases the nucleation rate due to its water-absorbing properties [52]. Amphiphilic polymers are preferred for hydrophobic carotenoids because the hydrophobic substituents of the excipients increase the interaction with carotenoids, and the hydrophilic substituents interact with water to enhance the dissolution [53]. In addition, lipids act as inhibitors or promoters of crystallization. The choice of lipids is based on three principles: hydrocarbon chain lengths, unsaturated degrees, and esterification degrees [54]. Saturated fatty acids with high polarity and a short chain length (e.g., butyric acid) have lower melting points, and the lower melting point prevents the molecule from forming crystalline nuclei. Meanwhile, fatty acids can adsorb on the interface of the liquid—solid nucleus to inhibit crystalline growth because of their surface-active properties [55].
Moisture also plays an essential role in crystalline formation. When water and APIs form crystals together, hydrates are produced, and the hydrogen bonding between water and APIs leads to higher lattice enthalpy and poorer bioavailability [5]. Hydroxyl groups of hydroxypropyl methylcellulose (HPMC) inhibit hydrate formation by occupying the sites where APIs and water have hydrogen bindings. The higher molecular weight of the excipients provides more functional groups for interactions and the full surface coverage of APIs [56]. Excipients with charges, such as dextran, alginate, and chitosan, can form ionic interactions with APIs possessing opposite charges, therefore, preventing the ongoing crystallization of APIs [56]. As excipients have great impacts on the stabilization of amorph conditions, excipient screening is an essential step for developing optimal formulations.

4.2. Preparation

Dissolution followed by rapid precipitation, melting followed by rapid cooling, and direct solid conversion are major methods to change the crystalline status of APIs. For the technique of dissolution followed by rapid precipitation, excipients dissolved in organic solvents loosen their structure, having interactions with the solvents. Meanwhile, APIs dissolved in organic solvents (i.e., amorphous states of APIs) enter the structure of the excipients and integrate into loosened excipients. With the rapid removal of solvents, the excipient and the amorphous APIs do not have enough time for ordinary crystallization and finally form a compact structure [57].
For the method of melting followed by rapid cooling, excipients and APIs are melted via heating. In the melt phase, the heat loosens the structure of the excipients, allowing APIs to occupy the space inside the excipients. Thereafter, fast cooling of the melt dramatically increases the viscosity and decreases the volume in a short time, resulting in much slower molecular mobility and molecule arrangement to prevent nucleation and crystalline growth [58].
Direct solid conversion refers to mechanical activation by milling. During the milling process, local heat accompanied by cooling results in amorphization, and the procedure increases static disorder and intrinsic dynamic disorder to the threshold value of the lattice, leading to the crumpling of the crystals. A limitation of this method, which should be taken into consideration, is the possibility of incomplete crystalline disorder [59]. The milling processing only renders the surface of the ingredients amorphized and may lead to inconsistent results in the physicochemical characterization [46].
In fact, for APIs, different manufacturing processes will produce varying degrees of amorphous forms, which cause diverse profiles in solubility, dissolution, and bioavailability [60].

4.3. Confinement (Change in Particle Sizes)

Confinement in the pharmaceutical field represents the physical restriction of APIs at the indicated scale. Under confinement, a different polymorph or amorph may appear, and confinement represents a practical handle to control or stabilize crystalline growth, as shown in Figure 7. The reasons why confinement affects the crystalline behavior remain unclear, but there are some proposed mechanisms: (i) When the size of the confinement is smaller than the critical nucleus size of the most stable crystals, the crystalline growth will be inhibited and a new polymorph or amorph will possibly form [61]. However, all polymorph forms of acetaminophen can grow under nanoconfinement [62]. The opposite result may relate to the chosen size for the confinement. Several pore sizes have been studied for the crystalline behavior of nifedipine, and McKenna and his colleague found a new polymorph presented at a certain pore size [63]. (ii) APIs in each compartment have to nucleate independently. As long as the compartment walls are not nucleated, homogeneous nucleation will dominate, which takes longer for crystallization. (iii) A thickness of 1 nm immobilizes the surface layer with high surface energy possibly forming at the compartment walls, and the immobilized layer slows the crystallization kinetics significantly [63,64]. The nanocompartments of the liposome truly inhibit crystalline growth [65]. Praziquantel occurs in its amorphous form under nanoconfinement because of its larger crystalline lattice, and amorphous praziquantel was found to increase the dissolution rate by five-fold [66]. This strategy can also be applied to hydrophobic carotenoids. Next, the application of these theories in carotenoid formulations will be introduced based on different types of formulations.

5. Approaches for the Crystalline Status Modification of Carotenoids

5.1. Co-Crystallization

Co-crystallization is defined as a single structurally homogeneous crystallization containing at least two neutral units (API and excipients) existing in solid and definite stoichiometric amounts [67]. This method is usually accomplished via supersaturation, which refers to slow cooling until the solubility limit is reached. Usually, the solubility of products prepared using this approach may not be significantly increased owing to the existence of a crystalline lattice structure, but it may provide several advantages, such as ease of preparation, lower hygroscopicity, and greater chemical stability of the products [68]. The commonly used excipients for co-crystallization systems should contain specific functional groups, including carboxylic acids (e.g., acetic acid and salicylic acid), amides (e.g., nicotinamide, saccharin, and urea), and alcohols (e.g., mannitol and sorbitol) to form intermolecular bonds with APIs [69].
The utilization of a supersaturated sucrose solution has been proposed for the preparation of carotenoid-rich extracts via the co-crystallization method [70], which aims to improve the dispersibility, hygroscopicity, and thermal stability of β-carotene. The ordered crystal of sucrose is transformed into an irregular and porous structure after the incorporation of β-carotene during the cooling and recrystallization processes. The crystalline status of co-crystallization can be evidenced by DSC and XRD examination. Though the technique of preparing pharmaceutical co-crystals with sucrose is believed to improve the solubility, dissolution, and other physicochemical properties of the encapsulated materials [71], merely the dissolution kinetics of sucrose have been determined in the current literature. One possible reason might be that the true solubility of the cocrystal products is not readily determined because API tends to transform into the most stable form in solution [72]. Few studies on carotenoid-loaded co-crystallization have discussed the crystalline state. Therefore, it may be an unexplored frontier and require more investigation for further discussion.

5.2. Solid Dispersion

Solid dispersion is a commonly used technology for crystalline state alternation and is defined as the dispersion of APIs in an inert carrier, such as sugars, polymers, and surfactants (Figure 8). Solvent evaporation and hot-melt methods are commonly operated, and the amorphous state may be produced during solvent removal or cooling [41]. The interaction between polymers and APIs generally results from the occurrence of hydrogen bonds and hydrophobic interaction. When solid dispersions are placed into an aqueous medium, such as simulated gastric fluid or simulated intestinal fluid, they would rapidly dissolve and exist in the supersaturation state owing to the amorphous state occurrence. Therefore, this may increase the aqueous solubility of poorly soluble APIs. Some polymers have been reported to retard crystalline growth in several ways: polyvinylpyrrolidone (PVP) suppresses the nucleation process and HPMC adsorbs on the surface of the crystal to prevent the formation of crystals [73]. In addition, the nucleation and growth procedures may be retarded via hydrogen bonding between APIs and excipients, which further inhibits crystalline formation [74].
It has been reported that β-carotene containing a solid dispersion composed of PVP and sucrose fatty acid ester (S-1670) was prepared by hot-melt extrusion and was found to be in an amorphous state via DSC and XRD. It was also proved to enhance solubility by about 390-fold, dissolution behavior, and also oral bioavailability [75,76,77]. A solid dispersion prepared using cyclic amylopectin has been used to protect β-carotene from light, heat and oxidation. The crystalline state of the solid dispersion was hard to detect using XRD owing to the uniform distribution in cyclic amylopectin. Starch was reported to inhibit the crystallization of a water-insoluble compound, β-carotene, and the composite was formed in an amorphous state. Cyclic amylopectin with a hydrophobic internal core could bind with β-carotene and hydrophobic compounds via intermolecular forces to generate a more amorphous formation [78].
Chang et al. [79] prepared lycopene dripping pills consisting of PEG 6000, Cremophor EL, and Tween 80 to improve the release behavior and oral bioavailability by approximately six-fold. The dripping pills were determined to be in an amorphous form via SEM and DSC. In this study, it was demonstrated that the lower viscosity caused by excessive emulsifiers may facilitate recrystallization.

5.3. Inclusion Complex

A complex is defined as the combination of APIs and ligands through hydrogen bonding, van der Waals forces, or hydrophobic effects [80]. Only a few compounds can be used as ligands to encapsulate hydrophobic carotenoids such as β-cyclodextrin, β-lactoglobulin, and amylose. The interaction of inclusion is shown in Figure 9.
Cyclodextrin is a cyclic oligosaccharide and is classified into different types based on the number of glucose residues, and it is the most well-studied complex ligand involving encapsulated carotenoids. Carotenoids can be encapsulated in the hydrophobic cavity of cyclodextrin through non-covalent interactions to stabilize the carotenoids with a random transformation from a crystalline to an amorphous state [84], and the hydrophilic outer surface of cyclodextrin helps the dissolution of carotenoids. Encapsulation in α-, β-, and γ-cyclodextrin was studied in tomato oil, which contained a lot of carotenoids. The complex presented as the microcrystalline in the emulsion form, and the complex powder was obtained with lyophilization to remove the solvent, accompanied by higher encapsulation and higher antioxidant capability [85]. The β-carotene/β-cyclodextrin complex was proposed, and β-carotene existed as an amorphous state in the complex, as shown in the results of DSC and XRD, leading to a 10- and 40-fold higher solubility and stability, respectively. Moreover, the antitumor activity was also improved [86]. The β-carotene/2-hydroxylproply-β-cyclodextrin/carrageenan/soy protein complex was also proposed and presented as an amorphous state in the DSC thermogram; it showed excellent bioaccessibility (78%) [87]. Astaxanthin has been prepared as a complex as well. The hexatomic side ring of astaxanthin was incorporated into the cavity of methyl-β-cyclodextrin to form a complex in an amorphous state, which was proved by the DSC thermogram, and the product exhibited 54-fold higher solubility, a 10-fold dissolution rate, and better bioaccessibility [88]. Sun et al. [81] proposed fucoxanthin (FX)/2-hydroxylpropyl-β-cyclodextrin via sonication and spray drying. The results of FTIR revealed that the characteristic peaks of FX disappeared, indicating that FX may have been successfully encapsulated, and the molecular docking suggested hydrogen bonding between FX and 2-hydroxylpropyl-β-cyclodextrin. XRD analysis also confirmed the amorphous state of FX. The FX/2-hydroxylpropyl-β-cyclodextrin complex showed better stability and antitumor activities toward HCT-116 and Caco-2 cells compared with free FX.
Amylose is also a food polymer that can accommodate hydrophobic carotenoids via V-amylose crystalline formation, and whether complexes are formed depends on the size, shape, and hydrophobicity of APIs. V-amylose is produced by the addition of ethanol as a precipitant; however, ethanol cannot incorporate into the hydrophobic cavity, and a hydrophobic carotenoid will be trapped by a nonspecific or specific interaction and stabilized with amylose polycrystals [89,90,91] which may be why APIs exist as amorphs in the amylose complex. However, among carotenoids, only β-carotene has been studied in the starch-complex system; the major finding was improved stability.
β-lactoglobulin is also regarded as a complex ligand for carotenoids, existing as the principal protein in whey protein. It can possibly bind hydrophobic carotenoids via the internal cavity of the β-barrel, the surface near Trp19-Arg124, and the groove between the α-helix and β-barrel of β-lactoglobulin [83,92]; the interaction may lower the molecular mobility to prevent recrystallization [93]. The binding mechanism has been studied in lycopene [94], but the effects on the bioactivity and physicochemical properties caused by the crystalline alteration still need to be further studied.

5.4. Micro/Nano Particles

Microparticle preparation includes spray drying, hot-melt extrusion, and phase separation [95], and the removal of solvents in spray drying as well as the cooling of the melt in extrusion causes APIs to remain in an amorphous state. In terms of phase separation, it works in a way similar to that of nanoprecipitation, which is the most common method used for nanoparticle preparation. The main concept of nanoprecipitation is solvent shifting, namely, the ouzo process. The schematic diagram of nanoprecipitation is shown in Figure 10, where hydrophobic APIs and polymers are dissolved in an organic solvent, and the organic solvent is added dropwise into an antisolvent (typically water). The solvent in the droplets moves toward the antisolvent, and the antisolvent does the reverse, causing a supersaturated state. This supersaturated state will further lead to the coprecipitation of hydrophobic APIs and the hydrophobic moiety of polymers forming nanoparticles with the surface coverage of the hydrophilic moiety of the polymers. The precipitated APIs remain amorphous as they do not have enough time to recrystallize in such a quick solvent replacement [96,97].
In our previous study about carotenoid nanoparticles [98], β-carotene-loaded PLGA-PVP nanoparticles were proposed, where amorphous β-carotene may benefit the solubility and oral absorption. Better oral absorption may result from nanoparticle morphology. Hu et al. [99] found that smooth and globular nanoparticles without irregularly lumpy astaxanthin crystals penetrate more easily into the cell. The theory has been applied in the micro-scale dimension, where astaxanthin-loaded hydrophilic microcapsules were obtained by spray drying. XRD analysis indicated that astaxanthin was encapsulated in an amorphous state, and the HepG2 cell growth inhibition activity was boosted [100]. Lutein and PVP have been prepared as particles to increase solubility and stability. PVP can inhibit crystallization by reducing molecular mobility, and the hydrogen bonding with lutein stabilizes the amorphous lutein, accompanied by higher stability against heat, light, and oxygen [101]. Lutein was also incorporated in zein nanoparticles and exhibited 80-fold higher water solubility. No crystalline peaks of lutein were found in XRD analysis as the nanoconfinement restricted the crystallization [102] and resulted in higher cellular uptake. Without excipients, the optical properties of β-carotene, lycopene, astaxanthin, and lutein nanoparticles obtained via nanoprecipitation were compared. With this preparation, the shell of the nanoparticles remained amorphous, and the core was still crystallized, as shown in cryo-TEM images. Moreover, the effective conjugation length of amorphous molecules was shorter than that of bulk crystals, and the absorption wavelength of amorphous molecules was blue-shifted. As a result, the color of the nanoparticles and crystals was rendered yellow and red, respectively [103]. In addition to the amorphous form, a new crystal may appear in the formulation. Ling et al. [104] proposed astaxanthin colloidal particles, and the decreased crystallinity led to a higher dissolution rate. Notably, astaxanthin has two common crystalline forms: polymorph I and polymorph II. A different polymorph was observed within the colloidal nanoparticle in the XRD analysis.

5.5. Lipid-Based Formulations

Lipid-based formulations, including emulsions, solid lipid nanoparticles (SLNs), nanostructured lipid carriers (NLCs), and self-emulsifying drug delivery systems (SEDDSs), are suitable for developing active lipid-soluble compounds, such as carotenoids, for oral bioavailability improvement. Emulsions, SLNs, and NLCs are composed of an aqueous phase and a lipid phase, with a surfactant for stabilization. The lipids used in these formulations are liquid, solid, and a mixture of liquid and solid oil, respectively (Figure 11). SEDDSs contain only oil and surfactants without water. Crystalline APIs would first be dissolved or melted to disperse in the oil to maintain the liquid state during the preparation procedure, so the APIs may be a solution type. Crystallization may occur after homogenization, cooling, or the storage period. In addition, supersaturation also causes the crystalline condition; therefore, the solubility of crystalline APIs in the solvent (solid lipid and liquid lipid) is crucial for crystallization. The general solubility equation (Equation (3)) is always utilized to calculate the solubility of crystalline APIs in a solvent using easily measurable properties [105].
Log Sw = 0.5 − 0.01 (Tm − 25) − logKOW
where SW is the molar water solubility, Tm is the melting point, and KOW is the oil–water partition coefficient of the solute.
For crystallization suppression, there are two main strategies: (i) enhancement of the saturated solubility to impede nucleation, and (ii) slowing down the diffusion for the prevention of crystalline growth (Figure 12). For the enhancement of the saturated solubility, the addition of surfactants, phospholipids, or high polarity, short-length saturated fatty acids could form micelles to reduce the driving force of nucleation and prevent crystal formation. Furthermore, the addition of non-polar agents, such as globular proteins or cyclodextrins, on their surfaces for incorporation with hydrophobic active compounds could also enhance the saturated solubility [55,105]. In order to slow down diffusion, viscosity changes and size reductions are often used. In a previous study, the addition of sugars enhanced the viscosity of the continuous phase to block the APIs’ diffusion, and the growth was retarded [105]. A previous study demonstrated that heterogeneous nucleations are confined when the average diameter of the drops is reduced and the crystallization is limited [54].
Apart from these strategies to prevent crystallization, the selection of lipids may affect the crystallization of imperfect crystalline or amorphous states and have different drug loadings, sizes, charges, and release behaviors [106]. The commonly used solid lipids in this formulation preparation contain triglycerides, waxes, fatty acids, and fatty alcohols and the lipid composition may have an influence on the crystalline state of SLNs/SLMs [107]. Taking the commonly used solid lipids, triglycerides as an example, these exhibit polymorphism upon cooling and possibly form the crystalline structure of α, β′, and β crystals with hexagonal, orthorhombic, and triclinic unit structures, respectively. The α-form is the most unstable structure, and spherical particles have been observed when triglycerides are in this form. During storage, the prepared formulation may spontaneously transfer the crystalline structure to a lower-energy state. Some lipids with higher polarity and amphiphilic properties, such as phospholipids, sterols, and di- and mono-acylglycerols, have been regarded as crystallization modifiers and affect the crystallization process [108]. In addition, the type of emulsifier may also affect the crystallization of lipid-based formulations [109]. The longer alkyl chain length of the surfactants has been shown to enhance the crystallinity of lipid-based formulations [107]. Thus, both the lipids and surfactants used in the formulation play a vital role in the modulation of the crystallization process. More research on each formulation is discussed as follows.

5.5.1. Solid Lipid Nanoparticles/Microparticles

The composition of solid lipid nanoparticles/microparticles (SLNs/SLMs) is similar to that of emulsions, but solid lipids are applied in the oil phase to have controlled release behavior or particle stability. The encapsulated drug may be prevented from crystallization and then form a solid solution. The distribution of carotenoids in SLNs may be uniform due to the high hydrophobicity and crystallization temperature [110]. High-energy-state α-crystals were found in the lipids of initial SLNs owing to rapid cooling, and the α-crystals may transfer to β-forms during storage [111]. Compared to α-crystals, highly lipophilic compounds such as carotenoids tend to be expelled under the β-crystalline state and the drug content may be reduced to affect the therapeutic effect. Therefore, the stability of SLNs is usually mentioned as a concern [112].
Some research has been reported about carotenoids containing SLNs/SLMs. β-carotene has been developed as an SLM using stearic acid and sunflower oil to prevent degradation during 7-month storage. The addition of sunflower oil resulted in less ordered crystals and induced an amorphous state, indicating the mixture of long-chained solid lipids and liquid lipids was suitable for the preparation of stable SLMs to prevent β-carotene degradation and exhibit excellent bioactivities [113]. Chen et al. [114] used both palm stearin and cholesterol as the solid lipid carrier of fucoxanthin to avoid the highly ordered crystalline structure of single solid lipids. The results showed that the SLN-microcapsules exist in an amorphous state owing to anti-solvent precipitation and ultrasonic treatment to form micelles easily and are capable of absorption by intestinal epithelial cells, indicating that the solubility of carotenoids could be enhanced to reduce the driving force of nucleation. The higher glass transition temperature indicated that the formulation has better temperature resistance [115]. Zeaxanthin was also prepared for SLNs using glycerol monostearate or glycerol distearate to resolve the problem of lipophilicity and instability. The crystallinity was determined to decrease in the formulation via the examination of DSC with the melting enthalpy decrease manner. The crystals of lipids preferred to form α-crystals with the high-energy state during the rapid cooling procedure [116]. Glycerol distearate is a mixture of C16 and C18 fatty acids and has a relatively low melting point and enthalpy compared to glycerol monostearate, indicating it has a poor crystalline structure. Therefore, SLNs prepared using glycerol distearate have the irregularity of the lipid crystals and display greater dissolution behavior [116].

5.5.2. Nanostructured Lipid Carrier

Nanostructured lipid carriers (NLCs) are similar to nanoemulsions and SLNs, but the lipids in NLCs include not only solid lipids but also liquid lipids. The incorporation of liquid lipids can allow the internal lipid phase to have a less-ordered crystalline arrangement to obviate the condition of active compound leakage and load more active compounds. Owing to the composition of both solid and liquid lipids, crystallinity is always considered. The crystalline index (CI) was reported to determine the crystalline state of APIs loaded in NLCs and it is calculated using Equation (4). A higher CI indicates that the encapsulation efficiency of NLCs may be higher owing to the less-ordered crystalline arrangement [106].
CI   ( % ) = M s M p   ×   γ   ×   100
where Ms indicates the melting enthalpy of NLCs, Mp indicates the melting enthalpy of pure solid lipids, and γ indicates the solid lipid concentration (%) in NLCs.
Astaxanthin-loaded NLCs have been developed to improve the physicochemical characteristics and storage stability. Glyceryl behenate and oleic acid were selected as solid and liquid lipids and lecithin and Tween 80 were chosen as surfactants in the oil phase and water phase, respectively. The authors evaluated the properties of NLCs prepared via lecithin removal, replacing Tween 80 with Tween 20 or replacing oleic acid with triacylglycerols, and the results showed that there is no improvement in the stability of the NLCs due to the chemically homogenous structure of the lipid mixture. In these formulations, β-crystals were formed and the aggregation, which contributed to the hydrophobic interaction, partial coalescence, or the penetration of lipid crystals, made the NLCs unstable. Among these solid lipids, NLCs prepared using glyceryl behenate were reported to have a more imperfect crystalline lattice and lead to high stability and entrapment. The crystal was demonstrated to be a metastable β′ polymorph, and the reduction in crystallinity compared to glyceryl behenate was determined by the broader and lower intensities in the XRD patterns and DSC thermogram. The incorporation of bioactive compounds may also make more imperfect crystals, leading to better encapsulation efficiency. The melting point of astaxanthin disappeared in the NLC group, indicating that astaxanthin is not in the crystalline state at this temperature and can be considered to be physically stable at high temperatures [115]. Oleic acid has been reported as a crystallization inhibitor. Oleic acid can adsorb and crystallize at the surface in the beginning so the crystallization may be hindered [54].
Glycerol monostearate or glycerol distearate as a solid lipid, medium-chain triglycerides as a liquid lipid, and soy lecithin and Tween 80 as surfactants were used to prepare NLCs to load zeaxanthin. The enthalpy reduction was observed in the NLC group compared to SLNs, indicating the crystallinity decrease in the lipid matrix. The lower enthalpy and crystallinity are capable of encapsulating more APIs and displaying better release behavior. Similar to the results of SLNs, NLCs prepared using glycerol distearate also had better properties. The results showed that the incorporation of liquid lipids and the selection of lipids have an influence on the crystallinity, and further improve the physicochemical characteristics of active compounds and formulations [116].

5.5.3. Microemulsion/Nanoemulsion

An emulsion is a mixture of two immiscible phases (aqueous phase and liquid oil phase) supported by a surfactant to reduce surface tension under thermodynamically unstable conditions. Solubilized and crystallized β-carotene nanoemulsions have been prepared to compare the influence of physical properties on bioaccessibility. The crystalline state was examined while passing through the stimulated digestion process, and no crystal was observed during digestion in the solubilized β-carotene nanoemulsion group. In the crystallized β-carotene nanoemulsion group, the initial crystals gradually disappeared and possibly contributed to the dilution by digestive juice in each step. The free fatty acid release profiles in the in vitro digestion study indicated that the physical state of β-carotene has no influence on lipid digestion. The bioaccessibility results showed that the solubilized β-carotene nanoemulsions had an 11.7- and 46-fold enhancement compared to crystallized β-carotene nanoemulsions and crystallized β-carotene in phosphate buffer saline, respectively, suggesting that the solubilization state without crystals is the suitable delivery strategy [117]. A previous study developed a lutein-loaded whey protein emulsion, which is similar to a Pickering emulsion. The crystalline form of the formulation containing only whey protein and phospholipids could be observed using a microscope, and the situation could be improved after the addition of mono- and di-glycerides. The mono- and di-glycerides benefited the solubility of lutein crystals and were demonstrated to be physical barriers in the crystalline growth process to prevent the carotenoids from crystallization, as well as improve the stability [118].

5.5.4. Self-Emulsifying Drug Delivery System

A self-emulsifying drug delivery system (SEDDS) is a mixture of active compounds, oils, surfactants, and co-surfactants through a gentle stirring procedure and the o/w emulsion is obtained by contact with digestion fluids and digestive motility. SEDDSs can be divided into two groups according to the droplet size. The droplet size of self-microemulsifying drug delivery systems and self-nanoemulsifying drug delivery systems is 100–250 nm and less than 100 nm, respectively [119]. A SEDDS is often in an amorphous state owing to being dissolved in lipids and surfactants. The crystallization always occurs during digestion due to the supersaturation to make excess active compounds precipitate or crystallize. In order to prevent supersaturation, some precipitation inhibitors have been reported, including cellulose (HPMC and hydroxypropyl cellulose), polymers (PVP and Soluplus®), surfactants (Tween, Cremophor, and D-α-Tocopherol polyethylene glycol 1000 succinate), and cyclodextrins [120,121]. The mechanism of the commonly used precipitation inhibitor HPMC, is to adsorb onto the surface to inhibit the nucleation and growth and to form intramolecular and intermolecular hydrogen bonds between the active compounds and itself via the hydroxyl groups in the structure. In addition, the “Spring and Parachute” is also observed in formulations with these excipients. Supersaturation may be rapidly reached, displaying spring behavior and the nucleation or crystalline growth may be restrained to stabilize the metastable supersaturated samples, witnessing the parachute phenomenon (Figure 13). In this way, the precipitation is inhibited, and there is a longer time for absorption in the expected regions [121]. The degree of supersaturation (S) is driven to precipitate, and it can be calculated using Equation (5) [122].
S =   total   drug   concentration saturation   concentration   of   the   drug   in   the   solvent
A SEDDS was incorporated with a solid dispersion, named a lipid-based solid dispersion (LBSD), to load lycopene to enhance the dissolution characteristics and oral absorption of lycopene. The results of XRD and DSC indicated that new signals appeared in the LBSD and that it had a lower melting point. The release behavior of the amorphous marketed product, Lycovit®, was significantly increased compared to the lycopene crystals, suggesting the benefit of crystalline transition. Although no obvious enhancement was observed in the LBSD owing to the non-amorphous state, the pharmacokinetic study demonstrated a significant improvement in oral absorption compared to Lycovit® due to the long-chained triglyceride for lymphatic transportation, suggesting the crystalline state did not fully affect the oral absorption in this study [123]. Aung et al. [124] prepared astaxanthin-loaded SMEDDS tablets containing SMEDDS, hydrophilic polymers as precipitation inhibitors, and microcrystalline cellulose for tableting. The SMEDDS, composed of rice bran oil, Kolliphor® RH 40, Span® 20, and two polymers, HPMC and polyvinyl alcohol, was used to obtain the supersaturation state and enhance the release of astaxanthin. The crystallinity was determined by PXRD and transferred from a crystalline to amorphous state after preparing the supersaturable SMEDDS. The SMEDDS with or without precipitation inhibitors enhanced the release behavior, antioxidant activity, and cellular uptake. The precipitation inhibitors in the SMEDDS could hinder the nucleation and precipitation and thus maintain astaxanthin in the solubilized form.

5.6. Liposome

The liposome is composed of a hydrophobic phospholipid bilayer shell and a hydrophilic core to form a spherical structure. Active lipophilic compounds tend to be encapsulated within the lipid bilayer and not precipitate in a crystalline form. Regarding the active hydrophilic compounds, they were loaded in the aqueous core with the crystalline precipitate, amorphous precipitate, or solution state depending on the properties of the active compounds and preparation process (Figure 14) [125]. Carotenoids are lipophilic compounds with high octanol–water partition coefficients; therefore, they are usually encapsulated within the phospholipid bilayer. Astaxanthin has been prepared for the liposome using the film dispersion-ultrasonic technique. XRD was conducted to examine the crystalline state of astaxanthin, soybean phosphatidylcholine (excipient), and the liposome. Pure astaxanthin was determined to be in a crystalline state owing to the existence of many peaks. The pattern of liposomes was different from that of astaxanthin and soybean phosphatidylcholine, implying that astaxanthin was successfully encapsulated in the liposomes with hydrogen bonds between astaxanthin and the phospholipid bilayer. The aqueous solubility of astaxanthin was observed to be enhanced 17-fold compared to pure astaxanthin, which may be attributed to the crystalline alternation of astaxanthin after liposome encapsulation [126].
Although there are some studies illustrating other factors majorly affecting oral absorption, such as intestine-specific transporters or lymphatic transport, solubility enhancement was mainly discussed and proved to efficiently improve oral bioavailability. The simplest way to enhance solubility is to alter the crystalline state from crystals to amorphs or polymorphs. The limitation is the instability of crystals owing to the high-energy state. To overcome this problem, there are many strategies and principles for crystallization suppression discussed in this article. To summarize, carotenoid formulations involved in the change of crystallinity are listed in Table 2. This article included various carotenoid-containing formulations, including those prepared using polymers or lipids, and the discussion of crystalline alternation. It may provide information to develop carotenoid-loaded formulations to deal with the problem of solubility and stability and exhibit outstanding bioactivities.

6. Conclusions

This review deals with the mechanisms of converting crystals into amorphs and stabilizing the amorphs in terms of polymer- and lipid-based formulations. Factors such as the types of excipients, manufacturing processes, and changes in particle size can transform the crystalline forms of APIs into other polymorph or amorph statuses. This review also provides representative and practical strategies for the delivery of carotenoids. These pharmaceutical technologies related to crystalline status modification efficiently improve the physicochemical properties of carotenoids, which amends their oral bioavailability and biological effects.

Author Contributions

Conceptualization, W.-Y.L., Y.-S.H., H.-H.K. and Y.-T.W.; methodology, W.-Y.L., Y.-S.H., H.-H.K. and Y.-T.W.; investigation, W.-Y.L. and Y.-S.H.; resources, W.-Y.L. and Y.-S.H.; writing—original draft preparation, W.-Y.L. and Y.-S.H.; writing—review and editing, W.-Y.L., Y.-S.H., H.-H.K. and Y.-T.W.; visualization, W.-Y.L. and Y.-S.H.; supervision, H.-H.K. and Y.-T.W.; project administration, H.-H.K. and Y.-T.W.; funding acquisition, Y.-T.W. W.-Y.L., H.-H.K. and Y.-S.H. contributed equally to this work. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Ministry of Science and Technology, Taiwan, R.O.C., under grant no. MOST110-2410-H-037-008-MY2. This study is supported partially by Kaohsiung Medical University Research Center Grant (KMU-TC111A04).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Thank Ting-An Liang for the image support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Gary Williamson, A.G.M.; Graham, A.B.; Catherine, S.B. Food Chemistry, Function and Analysis; Gary Williamson, A.G.M., Graham, A.B., Catherine, S.B., Eds.; RSC Publishing: Cambridge, UK, 2019; pp. 15–20. [Google Scholar] [CrossRef]
  2. Sridhar, K.; Inbaraj, B.S.; Chen, B.-H. Recent advances on nanoparticle based strategies for improving carotenoid stability and biological activity. Antioxidants 2021, 10, 713. [Google Scholar] [CrossRef] [PubMed]
  3. Stahl, W.; Sies, H. Bioactivity and protective effects of natural carotenoids. Biochim. Biophys. Acta 2005, 1740, 101–107. [Google Scholar] [CrossRef] [PubMed]
  4. Bayr, H. Reactive oxygen species. Crit. Care Med. 2005, 33, S498–S501. [Google Scholar] [CrossRef]
  5. Adejare, A. Remington: The Science and Practice of Pharmacy; Academic Press: Cambridge, MA, USA, 2020. [Google Scholar]
  6. Honda, M.; Kageyama, H.; Hibino, T.; Zhang, Y.; Diono, W.; Kanda, H.; Yamaguchi, R.; Takemura, R.; Fukaya, T.; Goto, M. Improved carotenoid processing with sustainable solvents utilizing Z-isomerization-induced alteration in physicochemical properties: A review and future directions. Molecules 2019, 24, 2149. [Google Scholar] [CrossRef] [PubMed]
  7. Stegemann, S.; Leveiller, F.; Franchi, D.; De Jong, H.; Lindén, H. When poor solubility becomes an issue: From early stage to proof of concept. Eur. J. Pharm. Sci. 2007, 31, 249–261. [Google Scholar] [CrossRef]
  8. Van den Mooter, G. The use of amorphous solid dispersions: A formulation strategy to overcome poor solubility and dissolution rate. Drug Discov. Today Technol. 2012, 9, e79–e85. [Google Scholar] [CrossRef]
  9. Uchiyama, K.; Naito, Y.; Hasegawa, G.; Nakamura, N.; Takahashi, J.; Yoshikawa, T. Astaxanthin protects β-cells against glucose toxicity in diabetic db/db mice. Redox Rep. 2002, 7, 290–293. [Google Scholar] [CrossRef]
  10. Park, J.S.; Chyun, J.H.; Kim, Y.K.; Line, L.L.; Chew, B.P. Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans. Nutr. Metab. 2010, 7, 1–10. [Google Scholar] [CrossRef]
  11. Wang, X.-j.; Chen, W.; Fu, X.-t.; Ma, J.-k.; Wang, M.-h.; Hou, Y.-j.; Tian, D.-c.; Fu, X.-y.; Fan, C.-d. Reversal of homocysteine-induced neurotoxicity in rat hippocampal neurons by astaxanthin: Evidences for mitochondrial dysfunction and signaling crosstalk. Cell Death Discov. 2018, 4, 1–11. [Google Scholar] [CrossRef] [Green Version]
  12. Wood, S.M.; Beckham, C.; Yosioka, A.; Darban, H.; Watson, R.R. β-Carotene and selenium supplementation enhances immune response in aged humans. Integr. Med. Int. 2000, 2, 85–92. [Google Scholar] [CrossRef]
  13. Yusuf, M.; Khan, R.A.; Khan, M.; Ahmed, B. Plausible antioxidant biomechanics and anticonvulsant pharmacological activity of brain-targeted β-carotene nanoparticles. Int. J. Nanomed. 2012, 7, 4311. [Google Scholar]
  14. Jayappriyan, K.; Rajkumar, R.; Venkatakrishnan, V.; Nagaraj, S.; Rengasamy, R. In vitro anticancer activity of natural β-carotene from Dunaliella salina EU5891199 in PC-3 cells. Biomed. Prev. Nutr. 2013, 3, 99–105. [Google Scholar] [CrossRef]
  15. Hira, S.; Saleem, U.; Anwar, F.; Sohail, M.F.; Raza, Z.; Ahmad, B. β-Carotene: A natural compound improves cognitive impairment and oxidative stress in a mouse model of streptozotocin-induced Alzheimer’s disease. Biomolecules 2019, 9, 441. [Google Scholar] [CrossRef]
  16. Takayanagi, K.; Morimoto, S.-i.; Shirakura, Y.; Mukai, K.; Sugiyama, T.; Tokuji, Y.; Ohnishi, M. Mechanism of visceral fat reduction in Tsumura Suzuki obese, diabetes (TSOD) mice orally administered β-cryptoxanthin from Satsuma mandarin oranges (Citrus unshiu Marc). J. Agric. Food Chem. 2011, 59, 12342–12351. [Google Scholar] [CrossRef]
  17. Heo, S.-J.; Jeon, Y.-J. Protective effect of fucoxanthin isolated from Sargassum siliquastrum on UV-B induced cell damage. J. Photochem. Photobiol. B Biol. 2009, 95, 101–107. [Google Scholar] [CrossRef]
  18. Maeda, H.; Hosokawa, M.; Sashima, T.; Murakami-Funayama, K.; Miyashita, K. Anti-obesity and anti-diabetic effects of fucoxanthin on diet-induced obesity conditions in a murine model. Mol. Med. Rep. 2009, 2, 897–902. [Google Scholar] [CrossRef] [PubMed]
  19. Woo, M.N.; Jeon, S.M.; Shin, Y.C.; Lee, M.K.; Kang, M.A.; Choi, M.S. Anti-obese property of fucoxanthin is partly mediated by altering lipid-regulating enzymes and uncoupling proteins of visceral adipose tissue in mice. Mol. Nutr. Food Res. 2009, 53, 1603–1611. [Google Scholar] [CrossRef]
  20. Palombo, P.; Fabrizi, G.; Ruocco, V.; Ruocco, E.; Fluhr, J.; Roberts, R.; Morganti, P. Beneficial long-term effects of combined oral/topical antioxidant treatment with the carotenoids lutein and zeaxanthin on human skin: A double-blind, placebo-controlled study. Ski. Pharmacol. Physiol. 2007, 20, 199–210. [Google Scholar] [CrossRef]
  21. Yu, C.-C.; Nandrot, E.F.; Dun, Y.; Finnemann, S.C. Dietary antioxidants prevent age-related retinal pigment epithelium actin damage and blindness in mice lacking αvβ5 integrin. Free Radic. Biol. Med. 2012, 52, 660–670. [Google Scholar] [CrossRef]
  22. Hammond Jr, B.R.; Miller, L.S.; Bello, M.O.; Lindbergh, C.A.; Mewborn, C.; Renzi-Hammond, L.M. Effects of lutein/zeaxanthin supplementation on the cognitive function of community dwelling older adults: A randomized, double-masked, placebo-controlled trial. Front. Aging Neurosci. 2017, 9, 254. [Google Scholar] [CrossRef]
  23. Sung, W.-S.; Lee, I.-S.; Lee, D.-G. Damage to the cytoplasmic membrane and cell death caused by lycopene in Candida albicans. J. Microbiol. Biotechnol. 2007, 17, 1797–1804. [Google Scholar]
  24. Qu, M.; Jiang, Z.; Liao, Y.; Song, Z.; Nan, X. Lycopene prevents amyloid [beta]-induced mitochondrial oxidative stress and dysfunctions in cultured rat cortical neurons. Neurochem. Res. 2016, 41, 1354–1364. [Google Scholar] [CrossRef]
  25. Hwang, S.; Lim, J.W.; Kim, H. Inhibitory effect of lycopene on amyloid-β-induced apoptosis in neuronal cells. Nutrients 2017, 9, 883. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, J.; Li, L.; Wang, Z.; Cui, Y.; Tan, X.; Yuan, T.; Liu, Q.; Liu, Z.; Liu, X. Supplementation of lycopene attenuates lipopolysaccharide-induced amyloidogenesis and cognitive impairments via mediating neuroinflammation and oxidative stress. J. Nutr. Biochem. 2018, 56, 16–25. [Google Scholar] [CrossRef] [PubMed]
  27. Huang, C.; Gan, D.; Fan, C.; Wen, C.; Li, A.; Li, Q.; Zhao, J.; Wang, Z.; Zhu, L.; Lu, D. The secretion from neural stem cells pretreated with lycopene protects against tert-butyl hydroperoxide-induced neuron oxidative damage. Oxid. Med. Cell. Longev. 2018, 2018, 1–14. [Google Scholar] [CrossRef] [PubMed]
  28. Zhu, N.-W.; Yin, X.-L.; Lin, R.; Fan, X.-L.; Chen, S.-J.; Zhu, Y.-M.; Zhao, X.-Z. Possible mechanisms of lycopene amelioration of learning and memory impairment in rats with vascular dementia. Neural Regen. Res. 2020, 15, 332. [Google Scholar] [CrossRef]
  29. Chasse, G.A.; Chasse, K.P.; Kucsman, A.; Torday, L.L.; Papp, J.G. Conformational potential energy surfaces of a lycopene model. J. Mol. Struct.: THEOCHEM 2001, 571, 7–26. [Google Scholar] [CrossRef]
  30. Khoo, H.-E.; Prasad, K.N.; Kong, K.-W.; Jiang, Y.; Ismail, A. Carotenoids and their isomers: Color pigments in fruits and vegetables. Molecules 2011, 16, 1710–1738. [Google Scholar] [CrossRef]
  31. Zia-Ul-Haq, M.; Dewanjee, S.; Riaz, M. Carotenoids: Structure and Function in the Human Body; Springer: Berlin/Heidelberg, Germany, 2021. [Google Scholar]
  32. Jing, C.; Qun, X.; Rohrer, J. HPLC separation of all-trans-β-carotene and its iodine-induced isomers using a C30 column. Thermo Sci. 2012, 391, 2–6. [Google Scholar]
  33. Yang, C.; Fischer, M.; Kirby, C.; Liu, R.; Zhu, H.; Zhang, H.; Chen, Y.; Sun, Y.; Zhang, L.; Tsao, R. Bioaccessibility, cellular uptake and transport of luteins and assessment of their antioxidant activities. Food Chem. 2018, 249, 66–76. [Google Scholar] [CrossRef]
  34. Yang, C.; Hassan, Y.I.; Liu, R.; Zhang, H.; Chen, Y.; Zhang, L.; Tsao, R. Anti-inflammatory effects of different astaxanthin isomers and the roles of lipid transporters in the cellular transport of astaxanthin isomers in Caco-2 cell monolayers. J. Agric. Food Chem. 2019, 67, 6222–6231. [Google Scholar] [CrossRef] [PubMed]
  35. During, A.; Hussain, M.M.; Morel, D.W.; Harrison, E.H. Carotenoid uptake and secretion by CaCo-2 cells: β-carotene isomer selectivity and carotenoid interactions1. J. Lipid Res. 2002, 43, 1086–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Furr, H.C.; Clark, R.M. Intestinal absorption and tissue distribution of carotenoids. J. Nutr. Biochem. 1997, 8, 364–377. [Google Scholar] [CrossRef]
  37. Sato, H.; Matsushita, T.; Ueno, K.; Kikuchi, H.; Yamada, K.; Onoue, S. Oily Suspension of Micronized Crystalline Lutein for the Improvement of Dissolution and Oral Bioavailability. Food Sci. Technol. 2022, 2, 272–279. [Google Scholar] [CrossRef]
  38. Faller, B.; Desrayaud, S.; Berghausen, J.; Laisney, M.; Dodd, S. 4 How solubility influences bioavailability. Solubility Pharm. Chem. 2020, 113, 113–132. [Google Scholar]
  39. Pudipeddi, M.; Serajuddin, A.T. Trends in solubility of polymorphs. J. Pharm. Sci. 2005, 94, 929–939. [Google Scholar] [CrossRef]
  40. Kaushal, A.M.; Gupta, P.; Bansal, A.K. Amorphous drug delivery systems: Molecular aspects, design, and performance. Crit. Rev. Ther. Drug Carr. Syst. 2004, 21, 133–193. [Google Scholar] [CrossRef]
  41. Vasconcelos, T.; Marques, S.; das Neves, J.; Sarmento, B. Amorphous solid dispersions: Rational selection of a manufacturing process. Adv. Drug. Deliv. Rev. 2016, 100, 85–101. [Google Scholar] [CrossRef]
  42. Healy, A.M.; Worku, Z.A.; Kumar, D.; Madi, A.M. Pharmaceutical solvates, hydrates and amorphous forms: A special emphasis on cocrystals. Adv. Drug. Deliv. Rev. 2017, 117, 25–46. [Google Scholar] [CrossRef]
  43. Zhou, W.; Greer, H.F. What can electron microscopy tell us beyond crystal structures? Eur. J. Inorg. Chem. 2016, 2016, 941–950. [Google Scholar] [CrossRef]
  44. Stevenson, H.P.; Makhov, A.M.; Calero, M.; Edwards, A.L.; Zeldin, O.B.; Mathews, I.I.; Lin, G.; Barnes, C.O.; Santamaria, H.; Ross, T.M. Use of transmission electron microscopy to identify nanocrystals of challenging protein targets. Proc. Natl. Acad. Sci. USA 2014, 111, 8470–8475. [Google Scholar] [CrossRef]
  45. Nurit, T.-G. Minerals Observed by Scanning Electron Microscopy (SEM), Transmission Electron Microscopy (TEM) and High Resolution Transmission Electron Microscopy (HRTEM). In Electron Microscopy; IntechOpen: London, UK, 2022. [Google Scholar]
  46. Bruni, G.; Berbenni, V.; Sartor, F.; Milanese, C.; Girella, A.; Franchi, D.; Marini, A. Quantification methods of amorphous/crystalline fractions in high-energy ball milled pharmaceutical products. J. Therm. Anal. Calorim. 2012, 108, 235–241. [Google Scholar] [CrossRef]
  47. Leyva-Porras, C.; Cruz-Alcantar, P.; Espinosa-Solís, V.; Martínez-Guerra, E.; Piñón-Balderrama, C.I.; Compean Martínez, I.; Saavedra-Leos, M.Z. Application of differential scanning calorimetry (DSC) and modulated differential scanning calorimetry (MDSC) in food and drug industries. Polymers 2019, 12, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Yu, D.G.; Branford-White, C.; Li, L.; Wu, X.M.; Zhu, L.M. The compatibility of acyclovir with polyacrylonitrile in the electrospun drug-loaded nanofibers. J. Appl. Polym. Sci. 2010, 117, 1509–1515. [Google Scholar] [CrossRef]
  49. Shah, B.; Kakumanu, V.K.; Bansal, A.K. Analytical techniques for quantification of amorphous/crystalline phases in pharmaceutical solids. J. Pharm. Sci. 2006, 95, 1641–1665. [Google Scholar] [CrossRef]
  50. Kumar, S.; Rao, R. Analytical tools for cyclodextrin nanosponges in pharmaceutical field: A review. J. Incl. Phenom. Macrocycl. Chem. 2019, 94, 11–30. [Google Scholar] [CrossRef]
  51. Mura, P. Analytical techniques for characterization of cyclodextrin complexes in the solid state: A review. J. Pharm. Biomed. Anal. 2015, 113, 226–238. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, J.; Liu, Z.; Wu, H.; Cai, T. Effect of polymeric excipients on nucleation and crystal growth kinetics of amorphous fluconazole. Biomater. Sci. 2021, 9, 4308–4316. [Google Scholar] [CrossRef]
  53. Wilson, V.R.; Lou, X.; Osterling, D.J.; Stolarik, D.F.; Jenkins, G.J.; Nichols, B.L.; Dong, Y.; Edgar, K.J.; Zhang, G.G.; Taylor, L.S. Amorphous solid dispersions of enzalutamide and novel polysaccharide derivatives: Investigation of relationships between polymer structure and performance. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef]
  54. Bayard, M.; Cansell, M.; Leal-Calderon, F. Crystallization of emulsified anhydrous milk fat: The role of confinement and of minor compounds. A DSC Study. Food Chem. 2022, 373, 131605. [Google Scholar] [CrossRef]
  55. Bayard, M.; Kauffmann, B.; Vauvre, J.-M.; Leal-Calderon, F.; Cansell, M. Isothermal crystallization of anhydrous milk fat in presence of free fatty acids and their esters: From nanostructure to textural properties. Food Chem. 2022, 366, 130533. [Google Scholar] [CrossRef] [PubMed]
  56. Gift, A.D.; Southard, L.A.; Riesberg, A.L. Influence of polymeric excipient properties on crystal hydrate formation kinetics of caffeine in aqueous slurries. J. Pharm. Sci. 2012, 101, 1755–1762. [Google Scholar] [CrossRef] [PubMed]
  57. Teja, S.B.; Patil, S.P.; Shete, G.; Patel, S.; Bansal, A.K. Drug-excipient behavior in polymeric amorphous solid dispersions. J. Excip. Food Chem. 2016, 4, 1048. [Google Scholar]
  58. Iyer, R.; Petrovska Jovanovska, V.; Berginc, K.; Jaklič, M.; Fabiani, F.; Harlacher, C.; Huzjak, T.; Sanchez-Felix, M.V. Amorphous Solid Dispersions (ASDs): The Influence of Material Properties, Manufacturing Processes and Analytical Technologies in Drug Product Development. Pharmaceutics 2021, 13, 1682. [Google Scholar] [CrossRef] [PubMed]
  59. Einfalt, T.; Planinšek, O.; Hrovat, K. Methods of amorphization and investigation of the amorphous state. Acta Pharm. 2013, 63, 305–334. [Google Scholar] [CrossRef] [PubMed]
  60. Noonan, T.J.; Chibale, K.; Cheuka, P.M.; Kumar, M.; Bourne, S.A.; Caira, M.R. Five solid forms of a potent imidazopyridazine antimalarial drug lead: A preformulation study. Cryst. Growth Des. 2019, 19, 4683–4697. [Google Scholar] [CrossRef]
  61. Diao, Y.; Whaley, K.E.; Helgeson, M.E.; Woldeyes, M.A.; Doyle, P.S.; Myerson, A.S.; Hatton, T.A.; Trout, B.L. Gel-induced selective crystallization of polymorphs. J. Am. Chem. Soc. 2012, 134, 673–684. [Google Scholar] [CrossRef] [PubMed]
  62. Beiner, M.; Rengarajan, G.; Pankaj, S.; Enke, D.; Steinhart, M. Manipulating the crystalline state of pharmaceuticals by nanoconfinement. Nano Lett. 2007, 7, 1381–1385. [Google Scholar] [CrossRef]
  63. Cheng, S.; McKenna, G.B. Nanoconfinement effects on the glass transition and crystallization behaviors of nifedipine. Mol. Pharm. 2019, 16, 856–866. [Google Scholar] [CrossRef]
  64. Rengarajan, G.; Enke, D.; Steinhart, M.; Beiner, M. Stabilization of the amorphous state of pharmaceuticals in nanopores. J. Mater. Chem. 2008, 18, 2537–2539. [Google Scholar] [CrossRef]
  65. Cipolla, D.; Wu, H.; Salentinig, S.; Boyd, B.; Rades, T.; Vanhecke, D.; Petri-Fink, A.; Rothin-Rutishauser, B.; Eastman, S.; Redelmeier, T. Formation of drug nanocrystals under nanoconfinement afforded by liposomes. RSC Adv. 2016, 6, 6223–6233. [Google Scholar] [CrossRef]
  66. Salas-Zúñiga, R.; Mondragón-Vásquez, K.; Alcalá-Alcalá, S.; Lima, E.; Höpfl, H.; Herrera-Ruiz, D.; Morales-Rojas, H. Nanoconfinement of a Pharmaceutical Cocrystal with Praziquantel in Mesoporous Silica: The Influence of the Solid Form on Dissolution Enhancement. Mol. Pharm. 2021, 19, 414–431. [Google Scholar] [CrossRef] [PubMed]
  67. Aakeröy, C.B.; Salmon, D.J. Building co-crystals with molecular sense and supramolecular sensibility. CrystEngComm 2005, 7, 439–448. [Google Scholar] [CrossRef]
  68. Steed, J.W. The role of co-crystals in pharmaceutical design. Trends Pharmacol. Sci. 2013, 34, 185–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Miroshnyk, I.; Mirza, S.; Sandler, N. Pharmaceutical co-crystals–an opportunity for drug product enhancement. Expert Opin. Drug Deliv. 2009, 6, 333–341. [Google Scholar] [CrossRef] [PubMed]
  70. Kaur, P.; Elsayed, A.; Subramanian, J.; Singh, A. Encapsulation of carotenoids with sucrose by co-crystallization: Physicochemical properties, characterization and thermal stability of pigments. LWT 2021, 140, 110810. [Google Scholar] [CrossRef]
  71. Chezanoglou, E.; Goula, A.M. Co-crystallization in sucrose: A promising method for encapsulation of food bioactive components. Trends Food Sci. Technol. 2021, 114, 262–274. [Google Scholar] [CrossRef]
  72. Good, D.J.; Rodriguez-Hornedo, N. Solubility advantage of pharmaceutical cocrystals. Cryst. Growth Des. 2009, 9, 2252–2264. [Google Scholar] [CrossRef]
  73. Huang, Y.; Dai, W.-G. Fundamental aspects of solid dispersion technology for poorly soluble drugs. Acta Pharm. Sin. B. 2014, 4, 18–25. [Google Scholar] [CrossRef]
  74. Newman, A.; Knipp, G.; Zografi, G. Assessing the performance of amorphous solid dispersions. J. Pharm. Sci. 2012, 101, 1355–1377. [Google Scholar] [CrossRef]
  75. Ishimoto, K.; Miki, S.; Ohno, A.; Nakamura, Y.; Otani, S.; Nakamura, M.; Nakagawa, S. β-Carotene solid dispersion prepared by hot-melt technology improves its solubility in water. J. Food Sci. Technol. 2019, 56, 3540–3546. [Google Scholar] [CrossRef] [PubMed]
  76. Otani, S.; Miki, S.; Nakamura, Y.; Ishimoto, K.; Ago, Y.; Nakagawa, S. Improved bioavailability of β-carotene by amorphous solid dispersion technology in rats. J. Nutr. Sci. Vitaminol. 2020, 66, 207–210. [Google Scholar] [CrossRef] [PubMed]
  77. Ishimoto, K.; Nakamura, Y.; Otani, S.; Miki, S.; Maeda, S.; Iwamoto, T.; Konishi, Y.; Ago, Y.; Nakagawa, S. Examination of dissolution ratio of β-carotene in water for practical application of β-carotene amorphous solid dispersion. J. Food Sci. Technol. 2022, 59, 114–122. [Google Scholar] [CrossRef] [PubMed]
  78. Wang, Y.; Chen, C.; Hu, X.; Campanella, O.H.; Miao, M. Fabrication and characterizations of cyclic amylopectin-based delivery system incorporated with β-carotene. Food Hydrocoll. 2022, 130, 107680. [Google Scholar] [CrossRef]
  79. Chang, C.-W.; Wang, C.-Y.; Wu, Y.-T.; Hsu, M.-C. Enhanced solubility, dissolution, and absorption of lycopene by a solid dispersion technique: The dripping pill delivery system. Powder Technol. 2016, 301, 641–648. [Google Scholar] [CrossRef]
  80. Fuenmayor, C.A.; Baron-Cangrejo, O.G.; Salgado-Rivera, P.A. Encapsulation of carotenoids as food colorants via formation of cyclodextrin inclusion complexes: A review. Polysaccharides 2021, 2, 454–476. [Google Scholar] [CrossRef]
  81. 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] [PubMed]
  82. Escobar-Puentes, A.A.; Reyes-López, S.Y.; Baltazar, Á.d.J.R.; López-Teros, V.; Wall-Medrano, A. Molecular interaction of β-carotene with sweet potato starch: A bleaching-restitution assay. Food Hydrocoll. 2022, 127, 107522. [Google Scholar] [CrossRef]
  83. Gheonea, I.; Aprodu, I.; Râpeanu, G.; Stănciuc, N. Binding mechanisms between lycopene extracted from tomato peels and bovine β-lactoglobulin. J. Lumin. 2018, 203, 582–589. [Google Scholar]
  84. Kaur, M.; Bawa, M.; Singh, M. β-Carotene-β-cyclodextrin inclusion complex: Towards enhanced aqueous solubility. J. Glob. Biosci. 2016, 5, 3665–3675. [Google Scholar]
  85. Durante, M.; Milano, F.; De Caroli, M.; Giotta, L.; Piro, G.; Mita, G.; Frigione, M.; Lenucci, M.S. Tomato oil encapsulation by α-, β-, and γ-Cyclodextrins: A comparative study on the formation of supramolecular structures, antioxidant activity, and carotenoid stability. Foods 2020, 9, 1553. [Google Scholar] [CrossRef] [PubMed]
  86. Yazdani, M.; Tavakoli, O.; Khoobi, M.; Wu, Y.S.; Faramarzi, M.A.; Gholibegloo, E.; Farkhondeh, S. Beta-carotene/cyclodextrin-based inclusion complex: Improved loading, solubility, stability, and cytotoxicity. J. Incl. Phenom. Macrocycl. Chem. 2022, 102, 55–64. [Google Scholar] [CrossRef]
  87. Lakshmanan, M.; Moses, J.A.; Chinnaswamy, A. Encapsulation of β-carotene in 2-hydroxypropyl-β-cyclodextrin/carrageenan/soy protein using a modified spray drying process. Int. J. Food Sci. Technol. 2022, 57, 2680–2688. [Google Scholar] [CrossRef]
  88. Nalawade, P.; Gajjar, A. Assessment of in-vitro bio accessibility and characterization of spray dried complex of astaxanthin with methylated betacyclodextrin. J. Incl. Phenom. Macrocycl. Chem. 2015, 83, 63–75. [Google Scholar] [CrossRef]
  89. Kim, J.-Y.; Seo, T.-R.; Lim, S.-T. Preparation of aqueous dispersion of β-carotene nano-composites through complex formation with starch dextrin. Food Hydrocoll. 2013, 33, 256–263. [Google Scholar] [CrossRef]
  90. Kong, L.; Bhosale, R.; Ziegler, G.R. Encapsulation and stabilization of β-carotene by amylose inclusion complexes. Food Res. Int. 2018, 105, 446–452. [Google Scholar] [CrossRef]
  91. Kim, J.-Y.; Huber, K.C. Preparation and characterization of corn starch-β-carotene composites. Carbohydr. Polym. 2016, 136, 394–401. [Google Scholar] [CrossRef]
  92. Allahdad, Z.; Khammari, A.; Karami, L.; Ghasemi, A.; Sirotkin, V.A.; Haertlé, T.; Saboury, A.A. Binding studies of crocin to β-Lactoglobulin and its impacts on both components. Food Hydrocoll. 2020, 108, 106003. [Google Scholar] [CrossRef]
  93. Buszewski, B.; Rodzik, A.; Railean-Plugaru, V.; Sprynskyy, M.; Pomastowski, P. A study of zinc ions immobilization by β-lactoglobulin. Colloids. Surf. A. Physicochem. Eng. Asp. 2020, 591, 124443. [Google Scholar] [CrossRef]
  94. Wang, C.; Chen, L.; Lu, Y.; Liu, J.; Zhao, R.; Sun, Y.; Sun, B.; Cuina, W. pH-Dependent complexation between β-lactoglobulin and lycopene: Multi-spectroscopy, molecular docking and dynamic simulation study. Food Chem. 2021, 362, 130230. [Google Scholar] [CrossRef]
  95. Thakur, S.; Sharma, R.; Garg, T. A review on microparticles: Preparation techniques and evaluation. Pharma Innov. J. 2022, 11, 837–840. [Google Scholar]
  96. Zhu, Z. Effects of amphiphilic diblock copolymer on drug nanoparticle formation and stability. Biomaterials 2013, 34, 10238–10248. [Google Scholar] [CrossRef] [PubMed]
  97. Cordt, C.; Meckel, T.; Geissler, A.; Biesalski, M. Entrapment of hydrophobic biocides into cellulose acetate nanoparticles by nanoprecipitation. Nanomaterials 2020, 10, 2447. [Google Scholar] [CrossRef] [PubMed]
  98. Liu, W.-Y.; Hsieh, Y.-S.; Wu, Y.-T. Poly (Lactic-Co-Glycolic) Acid–Poly (Vinyl Pyrrolidone) Hybrid Nanoparticles to Improve the Efficiency of Oral Delivery of β-Carotene. Pharmaceutics 2022, 14, 637. [Google Scholar] [CrossRef] [PubMed]
  99. Hu, F.; Liu, W.; Yan, L.; Kong, F.; Wei, K. Optimization and characterization of poly (lactic-co-glycolic acid) nanoparticles loaded with astaxanthin and evaluation of anti-photodamage effect in vitro. R. Soc. Open Sci. 2019, 6, 191184. [Google Scholar] [CrossRef]
  100. Nalawade, P.; Gajjar, A. Optimization of astaxanthin microencapsulation in hydrophilic carriers using response surface methodology. Arch. Pharm. Res. 2015, 1–17. [Google Scholar] [CrossRef]
  101. Zhao, C.; Cheng, H.; Jiang, P.; Yao, Y.; Han, J. Preparation of lutein-loaded particles for improving solubility and stability by Polyvinylpyrrolidone (PVP) as an emulsion-stabilizer. Food Chem. 2014, 156, 123–128. [Google Scholar] [CrossRef]
  102. Yuan, Y.; Li, H.; Liu, C.; Zhang, S.; Xu, Y.; Wang, D. Fabrication and characterization of lutein-loaded nanoparticles based on zein and sophorolipid: Enhancement of water solubility, stability, and bioaccessibility. J. Agric. Food Chem. 2019, 67, 11977–11985. [Google Scholar] [CrossRef]
  103. Suzuki, R.; Yasuhara, K.; Deguchi, S. Effect of Molecular Distortion on the Optical Properties of Carotenoid-Based Nanoparticles. J. Phys. Chem. C 2022, 126, 2607–2613. [Google Scholar] [CrossRef]
  104. Anarjan, N.; Tan, C.P.; Nehdi, I.A.; Ling, T.C. Colloidal astaxanthin: Preparation, characterisation and bioavailability evaluation. Food Chem. 2012, 135, 1303–1309. [Google Scholar] [CrossRef] [PubMed]
  105. McClements, D.J. Crystals and crystallization in oil-in-water emulsions: Implications for emulsion-based delivery systems. Adv. Colloid Interface Sci. 2012, 174, 1–30. [Google Scholar] [CrossRef] [PubMed]
  106. Chauhan, I.; Yasir, M.; Verma, M.; Singh, A.P. Nanostructured lipid carriers: A groundbreaking approach for transdermal drug delivery. Adv. Pharm. Bull. 2020, 10, 150. [Google Scholar] [CrossRef] [PubMed]
  107. Doktorovova, S.; Shegokar, R.; Souto, E.B. Role of excipients in formulation development and biocompatibility of lipid nanoparticles (SLNs/NLCs). In Nanostructures for Novel Therapy; Elsevier: Amsterdam, The Netherlands, 2017; pp. 811–843. [Google Scholar]
  108. Ribeiro, A.P.B.; Masuchi, M.H.; Miyasaki, E.K.; Domingues, M.A.F.; Stroppa, V.L.Z.; de Oliveira, G.M.; Kieckbusch, T.G. Crystallization modifiers in lipid systems. J. Food Sci. Technol. 2015, 52, 3925–3946. [Google Scholar] [CrossRef]
  109. Weiss, J.; Decker, E.A.; McClements, D.J.; Kristbergsson, K.; Helgason, T.; Awad, T. Solid lipid nanoparticles as delivery systems for bioactive food components. Food Biophys. 2008, 3, 146–154. [Google Scholar] [CrossRef]
  110. Gordillo-Galeano, A.; Mora-Huertas, C.E. Solid lipid nanoparticles and nanostructured lipid carriers: A review emphasizing on particle structure and drug release. Eur. J. Pharm. Biopharm. 2018, 133, 285–308. [Google Scholar] [CrossRef] [PubMed]
  111. Azizi, M.; Li, Y.; Kaul, N.; Abbaspourrad, A. Study of the physicochemical properties of fish oil solid lipid nanoparticle in the presence of palmitic acid and quercetin. J. Agric. Food Chem. 2019, 67, 671–679. [Google Scholar] [CrossRef] [PubMed]
  112. Helgason, T.; Awad, T.S.; Kristbergsson, K.; Decker, E.A.; McClements, D.J.; Weiss, J. Impact of surfactant properties on oxidative stability of β-carotene encapsulated within solid lipid nanoparticles. J. Agric. Food Chem. 2009, 57, 8033–8040. [Google Scholar] [CrossRef]
  113. Gomes, G.V.d.L.; Borrin, T.R.; Cardoso, L.P.; Souto, E.; Pinho, S.C.d. Characterization and shelf life of β-carotene loaded solid lipid microparticles produced with stearic acid and sunflower oil. Braz Arch Biol Technol. 2013, 56, 663–671. [Google Scholar] [CrossRef]
  114. Chen, Y.; He, N.; Yang, T.; Cai, S.; Zhang, Y.; Lin, J.; Huang, M.; Chen, W.; Zhang, Y.; Hong, Z. Fucoxanthin Loaded in Palm Stearin-and Cholesterol-Based Solid Lipid Nanoparticle-Microcapsules, with Improved Stability and Bioavailability In Vivo. Mar. Drugs 2022, 20, 237. [Google Scholar] [CrossRef]
  115. Tamjidi, F.; Shahedi, M.; Varshosaz, J.; Nasirpour, A. Design and characterization of astaxanthin-loaded nanostructured lipid carriers. Innov. Food Sci. Emerg. Technol. 2014, 26, 366–374. [Google Scholar] [CrossRef]
  116. Osanlou, R.; Emtyazjoo, M.; Banaei, A.; Hesarinejad, M.A.; Ashrafi, F. Preparation of solid lipid nanoparticles and nanostructured lipid carriers containing zeaxanthin and evaluation of physicochemical properties. Colloids. Surf., A. Physicochem. Eng. Asp. 2022, 641, 128588. [Google Scholar] [CrossRef]
  117. Xia, Z.; McClements, D.J.; Xiao, H. Influence of physical state of β-carotene (crystallized versus solubilized) on bioaccessibility. J. Agric. Food Chem. 2015, 63, 990–997. [Google Scholar] [CrossRef] [PubMed]
  118. Zhang, Y.; Yang, Y.; Mao, Y.; Zhao, Y.; Li, X.; Hu, J.; Li, Y. Effects of mono-and di-glycerides/phospholipids (MDG/PL) on the bioaccessibility of lipophilic nutrients in a protein-based emulsion system. Food Funct. 2022, 13, 8168–8178. [Google Scholar] [CrossRef] [PubMed]
  119. Tang, B.; Cheng, G.; Gu, J.-C.; Xu, C.-H. Development of solid self-emulsifying drug delivery systems: Preparation techniques and dosage forms. Drug Discov. Today 2008, 13, 606–612. [Google Scholar] [CrossRef]
  120. Kim, M.-S.; Ha, E.-S.; Choo, G.-H.; Baek, I.-H. Preparation and in vivo evaluation of a dutasteride-loaded solid-supersaturatable self-microemulsifying drug delivery system. Int. J. Mol. Sci. 2015, 16, 10821–10833. [Google Scholar] [CrossRef]
  121. Dokania, S.; Joshi, A.K. Self-microemulsifying drug delivery system (SMEDDS)–challenges and road ahead. Drug Deliv. 2015, 22, 675–690. [Google Scholar] [CrossRef]
  122. Quan, G.; Niu, B.; Singh, V.; Zhou, Y.; Wu, C.-Y.; Pan, X.; Wu, C. Supersaturable solid self-microemulsifying drug delivery system: Precipitation inhibition and bioavailability enhancement. Int. J. Nanomed. 2017, 12, 8801. [Google Scholar] [CrossRef] [Green Version]
  123. Faisal, W.; Ruane-O’Hora, T.; O’Driscoll, C.M.; Griffin, B.T. A novel lipid-based solid dispersion for enhancing oral bioavailability of Lycopene–In vivo evaluation using a pig model. Int. J. Pharm. 2013, 453, 307–314. [Google Scholar] [CrossRef]
  124. Aung, W.T.; Khine, H.E.E.; Chaotham, C.; Boonkanokwong, V. Production, physicochemical investigations, antioxidant effect, and cellular uptake in Caco-2 cells of the supersaturable astaxanthin self-microemulsifying tablets. Eur. J. Pharm. Sci. 2022, 176, 106263. [Google Scholar] [CrossRef]
  125. Li, T.; Cipolla, D.; Rades, T.; Boyd, B.J. Drug nanocrystallisation within liposomes. J. Control. Release 2018, 288, 96–110. [Google Scholar] [CrossRef]
  126. Pan, L.; Wang, H.; Gu, K. Nanoliposomes as vehicles for astaxanthin: Characterization, in vitro release evaluation and structure. Molecules 2018, 23, 2822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The structure of carotenes and xanthophylls.
Figure 1. The structure of carotenes and xanthophylls.
Pharmaceutics 15 00485 g001
Figure 2. The structures of E (trans)- and Z (cis)- forms of β-carotene.
Figure 2. The structures of E (trans)- and Z (cis)- forms of β-carotene.
Pharmaceutics 15 00485 g002
Figure 3. The difference among amorphism, polymorphism, and pseudo-polymorphism.
Figure 3. The difference among amorphism, polymorphism, and pseudo-polymorphism.
Pharmaceutics 15 00485 g003
Figure 4. PXRD profiles of β-carotene and its nanoformulations.
Figure 4. PXRD profiles of β-carotene and its nanoformulations.
Pharmaceutics 15 00485 g004
Figure 5. DSC thermograms of β-carotene, a β-carotene-PLGA-PVP physical mixture, and β-carotene-loaded PLGA-PVP nanoparticles.
Figure 5. DSC thermograms of β-carotene, a β-carotene-PLGA-PVP physical mixture, and β-carotene-loaded PLGA-PVP nanoparticles.
Pharmaceutics 15 00485 g005
Figure 6. Formulation conditions affecting crystallization.
Figure 6. Formulation conditions affecting crystallization.
Pharmaceutics 15 00485 g006
Figure 7. The concept of confinement and the relationship between confinement and crystals.
Figure 7. The concept of confinement and the relationship between confinement and crystals.
Pharmaceutics 15 00485 g007
Figure 8. The scheme of solid dispersions.
Figure 8. The scheme of solid dispersions.
Pharmaceutics 15 00485 g008
Figure 9. Scheme diagrams of (A) cyclodextrin, (B) amylose, and (C) β-lactoglobulin inclusion complexes [81,82,83].
Figure 9. Scheme diagrams of (A) cyclodextrin, (B) amylose, and (C) β-lactoglobulin inclusion complexes [81,82,83].
Pharmaceutics 15 00485 g009
Figure 10. The mechanism of trapping amorphous APIs in nanoparticles via nanoprecipitation [96].
Figure 10. The mechanism of trapping amorphous APIs in nanoparticles via nanoprecipitation [96].
Pharmaceutics 15 00485 g010
Figure 11. Illustration of a lipid-based formulation.
Figure 11. Illustration of a lipid-based formulation.
Pharmaceutics 15 00485 g011
Figure 12. The mechanism of crystallization suppression in a lipid-based formulation.
Figure 12. The mechanism of crystallization suppression in a lipid-based formulation.
Pharmaceutics 15 00485 g012
Figure 13. The mechanism of the “Spring and Parachute”.
Figure 13. The mechanism of the “Spring and Parachute”.
Pharmaceutics 15 00485 g013
Figure 14. Schematic of the hydrophilic and lipophilic compounds within a liposome. The right indicates that the hydrophilic compounds in the core may undergo crystallization and precipitation.
Figure 14. Schematic of the hydrophilic and lipophilic compounds within a liposome. The right indicates that the hydrophilic compounds in the core may undergo crystallization and precipitation.
Pharmaceutics 15 00485 g014
Table 2. The summary of carotenoid formulations involved in the crystalline change.
Table 2. The summary of carotenoid formulations involved in the crystalline change.
CarotenoidsFormulationCompositionCrystalline StatusResultsReference
CarotenoidsCo-crystalSucroseCrystalsThermal stability↑[70]
β-caroteneSolid dispersionPoly (vinyl pyrrolidone)
Sucrose fatty acid ester (S-1670)
AmorphsSolubility↑
Dissolution↑
Bioavailability↑
[75,76,77]
β-caroteneSolid dispersionCyclic amylopectionAmorphsStability↑[78]
β-carotγeneInclusion complexAmylose
(Amylomaize starch)
Both Amorphs and crystalsStability↑[89]
β-caroteneInclusion complexAmylose
(Corn starch)
Both Amorphs and crystalsStability↑[91]
β-caroteneInclusion complexAmylose
(High-amylose corn starch)
AmorphsStability↑[90]
β-caroteneInclusion complex2-hydroxylproply-β-cyclodextrin
Carrageenan
Soy protein
AmorphsBioaccessibility↑[87]
β-caroteneNanoparticlesPoly (lactic-co-glycolic) acid
Poly (vinyl pyrrolidone)
AmorphsBioavailability↑[98]
β-caroteneNanoemulsionCorn oilAmorphsBioaccessibility↑[117]
β-caroteneSolid lipid microparticlesStearic acid
Sunflower oil
Amorphs or
Less ordered crystals
Stability↑[113]
LycopeneSolid dispersion
(Dripping pills)
PEG 6000
Cremophor® EL
Tween® 80
AmorphsDissolution↑
Bioavailability↑
[79]
Lycopene
(Tomato oil)
Inclusion complexα, β, γ-cyclodextrinMicrocrystalsColor change
Stability↑
Antioxidation↑
[85]
LycopeneLipid based solid dispersionGelucire 44/14PolymorphsDissolution↑
Bioavailability↑
[123]
AstaxanthinInclusion complexMethyl-β-cyclodextrinAmorphsSolubility↑
Dissolution↑
Bioaccessibility↑
[88]
AstaxanthinColloidal particlesTween® 20
Sodium caseinate
Gum arabic
PolymorphsDissolution↑
Cellular uptake↑
[104]
AstaxanthinMicroparticlesPovidone K30
Copovidone
PEG 6000
Poloxamer 188
Tocopherol
Colloidal silicon dioxide
AmorphsHepG2 cell growth inhibition activity↑[100]
AstaxanthinNanoparticlesPoly(lactic-co-glycolic acid)AmorphsCellular uptake↑
Photoprotection↑
[99]
AstaxanthinLiposomeSoybean phosphatidyl choline
Cholesterol
Both Amorphs and crystalsSolubility↑
Stability↑
[126]
AstaxanthinNanostructured lipid carrierGlyceryl behenate
Oleic acid
Lecithin
Tween® 80
AmorphsStability↑[115]
AstaxanthinSelf-microemulsifying drug delivery systemRice bran oil
Kolliphor® RH 40
Span® 20
HPMC
Polyvinyl alcohol
AmorphsDissolution↑
Antioxidation↑
Cellular uptake↑
[124]
FucoxanthinInclusion complex2-hydroxylpropyl-β-cyclodextrinAmorphsSolubility↑
Stability↑
Anti-tumor activity↑
[81]
FucoxanthinSolid lipid nanoparticle-microcapsulesPalm stearin
Cholesterol
AmorphsSolubility↑
Stability↑
Bioavailability↑
[114]
LuteinParticlesPolyvinylpyrrolidone
Tween® 80
AmorphsStability↑[101]
LuteinNanoparticlesZein
Sophorolipid
AmorphsSolubility↑
Bioaccessibility↑
[102]
LuteinNanoemulsionBlending plant oil
Mono- and di-glycerides
Lecithin
Whey protein
AmorphsDissolution↑
Stability↑
[118]
ZeaxanthinSolid lipid nanoparticlesGlycerol monostearate
Glycerol distearate
Possible amorphsDissolution↑[116]
ZeaxanthinNanostructured lipid carrierGlycerol monostearate
Glycerol distearate
Medium-chain triglyceride Soy lecithin
Tween® 80
Possible amorphsDissolution↑[116]
↑: enhancement. ↓: reduction.
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

Liu, W.-Y.; Hsieh, Y.-S.; Ko, H.-H.; Wu, Y.-T. Formulation Approaches to Crystalline Status Modification for Carotenoids: Impacts on Dissolution, Stability, Bioavailability, and Bioactivities. Pharmaceutics 2023, 15, 485. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics15020485

AMA Style

Liu W-Y, Hsieh Y-S, Ko H-H, Wu Y-T. Formulation Approaches to Crystalline Status Modification for Carotenoids: Impacts on Dissolution, Stability, Bioavailability, and Bioactivities. Pharmaceutics. 2023; 15(2):485. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics15020485

Chicago/Turabian Style

Liu, Wan-Yi, Yun-Shan Hsieh, Horng-Huey Ko, and Yu-Tse Wu. 2023. "Formulation Approaches to Crystalline Status Modification for Carotenoids: Impacts on Dissolution, Stability, Bioavailability, and Bioactivities" Pharmaceutics 15, no. 2: 485. https://0-doi-org.brum.beds.ac.uk/10.3390/pharmaceutics15020485

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