Next Article in Journal
Assessment of the Substance Antioxidative Profile by Hyaluronan, Cu(II) and Ascorbate
Previous Article in Journal
Gibberellic Acid Initiates ER Stress and Activation of Differentiation in Cultured Human Immortalized Keratinocytes HaCaT and Epidermoid Carcinoma Cells A431
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Permeability of Buccal Mucosa

Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, 231 Albert Sabin Way, MSB # 3005, Cincinnati, OH 45267, USA
*
Author to whom correspondence should be addressed.
Pharmaceutics 2021, 13(11), 1814; https://doi.org/10.3390/pharmaceutics13111814
Submission received: 24 September 2021 / Revised: 18 October 2021 / Accepted: 23 October 2021 / Published: 31 October 2021
(This article belongs to the Section Biopharmaceutics)

Abstract

:
The buccal mucosa provides an alternative route of drug delivery that can be more beneficial compared to other administration routes. Although numerous studies and reviews have been published on buccal drug delivery, an extensive review of the permeability data is not available. Understanding the buccal mucosa barrier could provide insights into the approaches to effective drug delivery and optimization of dosage forms. This paper provides a review on the permeability of the buccal mucosa. The intrinsic permeability coefficients of porcine buccal mucosa were collected. Large variability was observed among the published permeability data. The permeability coefficients were then analyzed using a model involving parallel lipoidal and polar transport pathways. For the lipoidal pathway, a correlation was observed between the permeability coefficients and permeant octanol/water partition coefficients (Kow) and molecular weight (MW) in a subset of the permeability data under specific conditions. The permeability analysis suggested that the buccal permeation barrier was less lipophilic than octanol. For the polar pathway and macromolecules, a correlation was observed between the permeability coefficients and permeant MW. The hindered transport analysis suggested an effective pore radius of 1.5 to 3 nm for the buccal membrane barrier.

1. Introduction

For effective drug delivery across a biological membrane, it is important to understand the transport behavior of the membrane for the drugs. The mechanisms of drug permeation across a biological membrane can be divided into passive, facilitated, and active transport. The effects of the metabolic barrier due to enzyme degradation and drug clearance from the tissue due to blood circulation and the lymphatic system can also be important. For example, effective drug clearance after tissue penetration is beneficial to systemic drug delivery but has a negative impact on local drug delivery. In general, the barrier property of a membrane due to passive transport can be described by its permeability coefficient.
Previous reviews have covered a variety of topics in buccal drug delivery. These review papers, which span the last several decades, include general overviews and updates of buccal drug delivery and its assessments [1,2,3,4,5,6,7], drug products utilizing this route of administration [8,9], and technologies to improve drug delivery via the buccal mucosa such as penetration enhancers, microadhesives, nanoparticles, and biopolymers [10,11,12,13,14,15,16,17,18]. In addition, buccal delivery of specific drugs and their clinical uses have been reviewed [19,20,21,22]. Review papers are also available on the topics of buccal delivery of macromolecules such as peptides, oligonucleotides, and vaccines [7,23,24,25,26]. Consequently, these topics are not the focus of the present review paper.
For the permeability of buccal mucosa, previous studies have analyzed the relationships between the permeability coefficients and physicochemical properties of drugs [27,28]. The effect of temperature and activation energy of membrane permeation were also investigated to understand the transport mechanism of buccal mucosa [29]. However, a comprehensive summary of buccal permeability data is not available in the literature. The present paper provides a review on the permeability of the buccal mucosa for drug delivery. Porcine buccal mucosa has been the most common tissue in buccal drug delivery studies and was the focus of this review. The permeability coefficients of the buccal mucosa without the influence of formulations or the use of penetration enhancers (i.e., membrane intrinsic permeability) were summarized in this paper. The effects of lipophilicity and molecular weight of the permeants on their penetration across the buccal mucosa were examined. Analyses were performed to provide insights into a possible quantitative structure permeability relationship of passive transport across the buccal membrane that could be valuable in future drug delivery development.

2. Buccal Mucosa and Drug Delivery

Before the examination of buccal membrane permeability, this section provides a brief review of the buccal mucosa and buccal drug delivery. The oral mucosa is a complex series of tissues lining the oral cavity. It consists of tissue layers such as stratified squamous epithelium, basement membrane, and supporting connective tissues underneath. The buccal mucosa, in addition to the sublingual and gingival mucosa, is part of the oral mucosa. It is the area inside the cheek and between the gums and lower lips with an average surface area of ~100 cm2 [30]. The buccal mucosa consists of the outer epithelium and basement membrane. The non-keratinized stratified squamous epithelium forms the outer buccal epithelium. It is composed of mostly phospholipids and also proteins in the form of tonofilament. The basal layer of the epithelium differentiates into replacement cells that are shed from the outermost buccal surface. The epithelium, due to its morphology and lipid structure, is considered as the major barrier for the penetration of most drugs in buccal delivery. The basement membrane is a continuous layer of extracellular material and generally not considered as the major barrier for drug delivery. After the drug penetrates the buccal epithelium, it enters the systemic circulation via the vascularized tissue and jugular vein.
Buccal drug administration provides an alternative route for systemic drug delivery. It has several advantages over the gastrointestinal (GI) route by bypassing the hepatic first-pass effect, avoiding interference from acidity and enzymes relative to the GI tract, providing ease of dosing with the accessibility of the oral cavity and ease of drug removal in the event of adverse reactions. There are a number of commercial buccal drug delivery dosage forms in the market such as buccal tablet, spray, mucoadhesive, sublingual lozenge, chewing gum, film, and oromucosal solution. The disadvantages of buccal drug delivery include the (a) barrier of the buccal mucosa that its permeability may be too low for certain drugs, (b) interference of saliva that can dilute the drug for absorption, and (c) variable environment in the oral cavity due to food consumption and other daily activities. For example, although the permeability of the buccal mucosa is generally higher than that of the stratum corneum [31], it is generally lower than that of the GI mucosal monolayer [32,33], as can be seen by comparing their values to the buccal permeabilities presented later in the present review. To overcome this barrier, chemical penetration enhancers such as surfactants, physical penetration enhancers such as iontophoresis, and formulation technologies such as mucoadhesive and polymeric dosage forms can be utilized for buccal drug delivery. In order to fully utilize these enhancement methods and to develop effective buccal drug delivery systems, it is important to understand the intrinsic passive permeability of the buccal mucosa.

3. Permeability of Buccal Mucosa and Data Variability

The permeants and their buccal permeability coefficients collected for this review are listed in Table A1 (see Appendix A). The permeability coefficients were obtained from the references [27,28,29,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107] and the logarithmic values of the permeability coefficients in cm/s (log P) were calculated. Only permeability data from porcine buccal mucosa were collected for the analysis because the majority of previous permeation studies was with the porcine tissue. In addition, a comparison between the porcine tissue and tissues from other species (bovine, canine, hamster, monkey, and human) showed that their permeability coefficients were within similar orders of magnitudes except for hamster tissue that displayed higher permeability (Table 1). In some cases, engineered tissues (TR146 tissue model, EpiOral tissue model, and Caco-2 cells) also showed higher permeability than the porcine buccal tissue. It should be noted that the data shown in Table 1 are not intended to be comprehensive but to support the use of porcine buccal permeability data in the analyses of the present review. In the data collection, another selection criterion was unperturbed buccal membrane. Only buccal permeability coefficients (intrinsic permeability) without formulation influences and organic solvent manipulations were considered. Results obtained at temperatures outside the normal range of 34–37 °C were not included. For the model analyses, the physicochemical properties of the permeants such as pKa, octanol/water partition coefficient (Kow), and octanol/water distribution coefficient (Dow), when available, were also collected from the previous studies (Table A1). Dow, which is the ratio of the concentration of unionized species in the octanol phase to the concentration of both ionized and unionized species in the aqueous phase, is related to Kow as described by Equation (A2) (see Appendix B). When the pKa, Kow, or Dow values were not available in the references, the pKa and Kow values were obtained from PubChem [108] and Dow was calculated using the pKa and pH of the permeant solutions in the studies (see Equations (A3) and (A4)).
When permeability data of the same permeants are available from buccal permeation studies performed by multiple research groups under similar experimental conditions, large variability was observed between the data generated among these research groups. To compare the permeability data in these studies (from different research groups), the average of the mean permeability coefficients presented in these studies was determined for the permeant and the coefficient of variation (CV) was calculated (CV = ratio of the standard deviation to the average × 100%). The CV of the permeability coefficients was then plotted against the average permeability coefficient of each permeant (Figure 1). Although the high CV from the n = 2 studies can be related to the nature that only two studies were compared, most permeants have CV much higher than 50% even without considering the variability of the n = 2 studies. This is significantly higher than the CV generally encountered in diffusion cell permeation experiments [119]. Possible causes of the large variability could be the (a) different sources of porcine tissues (age, species), (b) different methods of tissue preparation and the resulting tissue conditions, and (c) different experimental conditions among these studies. For comparison, the average and CV of the mean permeability coefficients from studies of the same groups (from different permeation studies such as different papers published by the same research group) were also calculated and the CV was plotted against the average permeability coefficient of each permeant in the figure. The CV values from the same research groups are generally smaller than those from different research groups (open symbols vs. closed symbols, respectively). Furthermore, there is no apparent relationship between the variabilities and permeability coefficients of the permeants in both comparisons (of the same and different research groups), indicating that the variabilities were likely not transport-mechanism related. The large variabilities observed among the permeability data of the same permeants from the studies published by different research groups suggested the potential difficulty in using the literature permeability data to determine the quantitative structure permeability relationship for buccal drug delivery.
For the permeability analyses in the present review, when the permeability data were from different studies of the same research group, the average value of the permeability coefficients published in these studies was calculated and used as an individual data point. When the permeability data were from different research groups, the result from each research group was treated as an individual data point in the analyses; these data were analyzed as separate data points with the same weighing factor as if they were different permeants.

4. Membrane Transport Theory

In order to examine the relationships between the permeability coefficients of buccal mucosa and the physicochemical properties of the permeants, this section provides a brief review of transport theory for drug permeation across biological membranes. In general, the permeability of a biological membrane can be described by a parallel pathway permeation model of lipoidal and polar pathways [120,121]. The lipoidal pathway (or transcellular pathway) describes the permeation across the lipid barrier from the membrane lipid lamella structure, and the polar pathway (or pore pathway) describes the permeation across the aqueous channels across the membrane via the paracellular route or membrane defects. For the permeation of a weak acid or weak base, which is pH dependent, the flux of the permeant and its permeability coefficient can be expressed as [32]:
J = J u + J i = P u C u + P i C i = P u f u + P i ( 1 f u ) C
P = P l + P p f u + P p ( 1 f u ) = P l f u + P p
where J is flux, P is permeability coefficient of the membrane, C is concentration, fu is the fraction of unionized permeant, subscripts u and i represent the unionized and ionized permeants, and subscripts l and p represent the lipoidal and polar pathways, respectively. Assuming that the microenvironment of the lipid lamellae in the membrane can be mimicked by octanol and the contribution of Pp is minimal, and using the relationship between membrane diffusion coefficient and permeant molecular weight [31], the permeability coefficient can be expressed as (see derivation of Equation (A10) in Appendix B):
log P = log D o w + c   M W + c o n s t a n t
where MW is permeant molecular weight, c is the coefficient of MW, and constant is a constant. For permeants that are not affected by pH, Dow = Kow and Equation (3) becomes:
log P = log K o w + c   M W + c o n s t a n t
Equations (3) and (4) were the models used in the analyses of the buccal permeability data (see Section 5).
When the microenvironment of the lipid barrier is different from that of octanol, membrane partitioning can be described by the linear free energy relationship [122]:
log K m = a log K o w + log b .  
where Km is membrane partition coefficient and a and b are constants. The slope of the log P vs. log Kow relationship (or log P vs. log Dow for the permeants that are pH dependent due to ionization) indicates the lipophilicity of the rate limiting barrier for permeation across the membrane. Replacing Kow in the derivation of Equation (3) by this free energy relationship, Equation (4) can be rewritten as (see the derivation of Equation (A13) in Appendix B):
log ( P / D o w ) = a 1 log K o w + c   M W + c o n s t a n t
In addition to Equations (3) and (4), the buccal permeability data were also analyzed by Equation (6) (see Section 5).
For permeation across the polar pathway in a biological membrane, which can be modeled as aqueous channels with hindered transport, the permeability coefficient of the polar pathway (Pp) can be described by the polar pathway transport model [123,124].
P p = ε K p H D a q / h m
where ε is membrane porosity, Kp is the partition coefficient due to permeant-to-membrane interactions, Daq is aqueous diffusion coefficient, H is hindered transport factor for diffusion, and hm is the effective thickness of the membrane for the polar pathway. The hindered transport factor is a function of the permeant molecular size and the pore size of the polar pathway [125].
H = 6 π 1 λ 2 / 2.25 π 2 2 1 λ 2.5 1 + n = 1 2 a n 1 λ n + 0 4 a n + 3 λ n
where λ is the ratio of permeant radius to pore radius, a1 = −1.21667, a2 = 1.53359, a3 = −22.5083, a4 = −5.6117, a5 = −0.3363, a6 = −1.216, and a7 = 1.647. When λ < 0.4, Equation (8) is equivalent to the commonly used Renkin equation. The hindered transport model can be used to characterize the effective size of a pore transport pathway [126,127,128]. Assuming that the effects due to permeant-to-membrane interactions in the aqueous transport pathway are small (i.e., Kp ≈ 1), the ratio of the permeability coefficients of two permeants is related to the ratio of hindered transport factors of the permeants.
P p , 1 / D a q , 1 / P p , 2 / D a q , 2 = H 1 / H 2
where subscripts 1 and 2 represent permeant 1 and permeant 2, respectively. By fitting Equation (9) with the permeability ratio data, the effective pore radius of the polar pathway can be evaluated (see Section 6).

5. Effects of Lipophilicity and Molecular Weight on the Permeation of Small Molecules

Figure 2 shows the relationship between the permeability coefficients of buccal mucosa and the lipophilicities of the permeants in Table A1, in which the lipophilicities are measured by Kow. The correlation between log P and log Kow was poor. By taking into account the fraction of ionization of the permeants, Figure 3 shows the relationship between the permeability coefficients and Dow of the permeants. The correlation between the permeability coefficients and lipophilicities of the permeants improved when Dow (instead of Kow) was used (Figure 3 vs. Figure 2), but the correlation was still relatively poor.
Figure 4 shows the relationship between the permeability coefficients of buccal mucosa and the MW of the permeants. There was no direct correlation between the permeability coefficients and MW of the permeants without the consideration of their lipophilicities. The effect of permeant MW on membrane permeability was then investigated by regression analyses (MS Excel Linest function) using log Dow and MW as two independent variables. Three parameters (a, c, and constant) were fitted to the experimental data using a similar relationship as Equation (3):
log P = a log D o w + c   M W + c o n s t a n t
Table 2 presents the result of this analysis. The incorporation of MW in the permeability analysis did not have any significant effects on the correlation between log P and log Dow for the quantitative structure permeability relationship.
Equation (10) was derived under the assumption of minimal contribution from the polar pathway to the total permeability of the buccal membrane (i.e., Pp << P; see the assumption in Equation (A8) in Appendix B). For example, permeants that have low Dow (e.g., Dow < −1) could predominantly utilize the polar pathway for buccal membrane permeation and have low or comparable permeability coefficients to those of the polar pathway, so the inclusion of these data in the analysis can introduce errors. To this end, log Dow < −1 was applied as an exclusion criterion in the evaluation of the “log P vs. log Dow and MW” relationship; only the permeability data of permeants with log Dow > −1 and water were analyzed. Water was not excluded in the analyses because it could readily permeate lipid bilayers without using the polar pathway. The exclusion of these data did not improve the “log P vs. log Dow and MW” correlation and further exclusion of the permeants that have Dow < 0 also did not show any improvement (Table 2). To account for possible differences between Km and Kow, additional analyses were performed using Equation (6) and the permeability data. The results are presented in Table 2. The approach of log (P/Dow) vs. log Kow and MW provided better correlations than those of the “log P vs. log Dow and MW” when compared using the same permeability datasets (first row vs. fourth row, second row vs. fifth row, and third row vs. sixth row in Table 2). However, except for the dataset of “all data” (fourth row in Table 2), the correlations were still relatively poor. The improvement in correlations using the model of Equation (6) is consistent with the conclusion that the buccal membrane barrier is less lipophilic than octanol (Km vs. Kow; see discussion later in this section and Section 7). For the correlation using all data, the quantitative structure permeability relationship observed was: log (P/Dow) = −0.56 log Kow −0.0014 MW −4.8 (with R2 = 0.588).
Membrane transport of weak acid and weak base can be affected by a number of factors including the fraction of permeant ionization that is a function of permeant pKa and solution pH (i.e., pH dependent permeant charges). Due to this uncertainty, permeant transport that is pH dependent and is related to the fraction of permeant ionization can introduce errors in the permeability analyses. Figure 5 shows the relationship between the permeability coefficients and Kow by excluding the permeants with ionization as a function of pH (i.e., using only pH independent permeants) in the analysis. The exclusion of the pH dependent permeants slightly improved the correlation of the relationship between log P and log Kow (compared to Figure 2).
With the pH independent permeants as the dataset, regression analyses were performed using log Kow and MW as two independent variables, a, c, and constant as the parameters, and a similar relationship as Equation (4):
log P = a log K o w + c   M W + c o n s t a n t
The incorporation of MW in the analysis improved the correlation (Table 3). To consider the influence of the polar pathway (Pp), the exclusion criteria of log Kow < −1 and < 0 were applied to the analyses of these permeants. The exclusion of the permeants that have log Kow < −1 provided a better correlation of the “log P vs. log Kow and MW” relationship. Increasing the log Kow value in the exclusion criterion to analyze only permeants with log Kow > 0 further improved the “log P vs. log Kow and MW” correlation (R2 = 0.615). The results of these analyses are summarized in Table 3.
An interesting observation in the present study is the slopes of log P vs. log Kow (or log P vs. log Dow) in the permeability, lipophilicity, and MW correlation analyses. These slopes (coefficient a) are significantly smaller than unity for the linear free energy relationship between membrane partitioning and octanol/water partitioning (Km vs. Kow, Equation (5)), and this can be attributed to a number of factors. First, the transport rate-limiting barrier of the buccal mucosa can be less lipophilic compared to octanol. This can lead to a slope significantly smaller than unity in the linear free energy relationship between water-to-membrane and water-to-octanol partitioning for buccal membrane permeation. Second, the influences of the polar pathway and underlying tissues of the buccal tissues can create lower and upper boundaries, respectively, for the buccal permeability range to accurately determine the slope (coefficient a) in the linear free energy relationship. When the permeability range is narrow in the permeability vs. lipophilicity relationship, the data points at the boundaries or outside this range can “skew” the data, resulting in an apparent slope less than the actual value in the “log P vs. log Kow and MW” (or “log P vs. log Dow and MW”) correlation. Although employing the exclusion criteria of log Kow (or log Dow) < −1 and < 0 could minimize the impact of the polar pathway and reduce this “skewing” effect in the data analysis, as shown in the improvement of the correlations in Table 3, the use of these exclusion criteria might not be sufficient. For example, there was no improvement in the correlation after applying the log Dow < −1 and < 0 exclusion criteria in the correlations in Table 2. Third, the variability of the permeability data in the previous studies and the uncertainties of log Dow can affect the correlations. Particularly, the general improvement of the “log P vs. log Kow and MW” correlations with the pH independent permeants (i.e., without changes in ionization such as fraction of ionization due to pH) over the correlations of “log P vs. log Dow and MW” (first three rows in Table 2 vs. Table 3) can be attributed to the decrease in uncertainties related to permeant ionization due to pH. Previous studies have suggested the possibility of membrane pH that is different from solution pH surrounding the biological membrane (e.g., skin, GI) [129,130,131], and this type of phenomena can lead to errors in calculating the fraction of ionization for buccal membrane permeation in the analyses. Regardless of these influencing factors and uncertainties in the present permeability analyses, the results indicate apparent correlation slopes of buccal membrane permeation vs. log Kow that are significantly smaller than those of other biological membranes such as lipid bilayers (~2.0–2.8), skin (~0.7–0.8), and cornea (~0.5) [31,122,132], except when the model of Equation (6) was used. In addition, there is a lack of molecular size (or MW) dependence for the permeation of small molecules across the lipoidal barrier of the buccal membrane, which may be “masked” by data variability, when all the data were included in the analyses. When the exclusion criteria were applied to the permeants to take into account the model limitations and the uncertainties in permeant pH-ionization and partitioning, a reasonable quantitative structure permeability relationship was observed: log P = 0.2 log Kow−0.0033 MW−4.8 (with R2 = 0.615, Table 3 last row).

6. Effective Pore Size for the Permeation of Macromolecules

The permeation of macromolecules across a biological membrane is anticipated to be through the paracellular route of the membrane. This transport pathway can be modeled by the polar pathway and hindered transport theory (Equations (7) and (8)). Figure 6a presents the relationship between the permeability coefficients and MW of the macromolecules and the polar permeants that have log Kow < −1 and are not pH dependent (i.e., not weak acid or weak base) for the buccal mucosa. With the inclusion of the macromolecules in the permeability analysis, unlike the analysis of only small permeants, a steep size dependence of permeability was observed with a linear regression slope of −1.58 and R2 = 0.613 for the log P vs. log MW relationship.
To examine the effective pore size of the polar pathway, the ratios of the permeability and diffusion coefficients were calculated and compared to a reference (Equation (9)). The reference point in this analysis (Permeant 2 in Equation (9)) was a hypothetical small permeant with an average permeability coefficient and MW of the small permeants that have MW < 300 Dalton in Figure 6a. Figure 6b shows the relationship between the P/Daq ratio and MW from the permeability data and the H ratio calculated using Equation (8), based on the reference point. The theoretical hindered transport relationship of molecular size and pore size shows a significant decrease in the H ratio when the molecular size increases from 300 to 20,000 Dalton and the pore size (radius) decreases from 4 nm to 1.5 nm. By comparing the theoretical hindered transport H ratios and the experimental P/Daq ratios, the result suggests an effective pore radius in the range of 1.5 to 3 nm for the buccal polar pathway (or paracellular pathway). The result in this analysis is in agreement with that in a previous study that investigated the polar pathway using a polymer of different molecular sizes (polyethylene glycol) [96]. In addition, the effective pore size of the buccal mucosa was in the same order of magnitude as other biological membranes such as skin, cornea, conjunctiva, nail, and GI mucosal monolayer [124,126,133,134].

7. Permeability Analysis Discussion and Consideration

The present review examined the permeability of porcine buccal mucosa and the major findings are as follows. Large variability of the permeability data was observed in previous studies from different research groups even when the permeability measurements were performed under similar experimental conditions (Figure 1). In general, the correlation between buccal membrane permeability and permeant lipophilicity was relatively poor (Figure 2) and no general permeability-to-MW relationship was observed (Figure 4) when either permeant lipophilicity (as indicated by Kow) or MW was used as the single independent variable in the analyses of the permeability data for all permeants.
To further investigate the relationship between permeant lipophilicity and the permeability of the lipoidal pathway in the buccal mucosa, the permeability was described better by log Dow than log Kow (Figure 3 vs. Figure 2) consistent with the transport theory of uncharged permeants due to pH dependent ionization of the permeants (i.e., weak acid and weak base) when all permeants were used in the analyses. However, the correlation between log P and log Dow was still relatively poor with no improvement in the correlation when permeant MW was incorporated as an additional independent variable in the analyses; there was no observable effect of permeant MW on the permeability. To take into account the contribution of the polar pathway (Pp) to membrane permeability, the permeability data were also evaluated by allowing only permeants with log Dow > −1 or > 0 in the analyses. These exclusion criteria did not improve the correlations of the “log P vs. log Dow and MW” relationship for the permeants studied (Table 2). A hypothesis of the poor correlations is data variability and errors related to the fraction of ionization calculations (e.g., membrane pH different from solution pH and errors introduced in the calculations of log Dow). The data were therefore analyzed using only pH independent permeants (permeants without fraction of ionization as a function of pH).
For the pH independent permeants, a better correlation was observed in the log P vs. log Kow relationship (Figure 5 vs. Figure 2). The correlation improved with the incorporation of MW as an additional independent variable and further improvement was observed with the exclusion of the permeants that likely utilize the polar pathway, i.e., by limiting the analyses to only permeants with log Kow > −1 and > 0 (Table 3). When these exclusion criteria were applied, a reasonable quantitative structure permeability relationship of “log P vs. log Kow and MW” was observed (R2 = 0.615). Another observation in the analyses is the coefficient of log Kow in the “log P vs. log Kow and MW” relationship (coefficient a in Equation (11)). This coefficient denotes the slope of the linear free energy relationship between Km and Kow (see Equation (5)). The slope of 0.2 is smaller than those observed with other biological membranes and suggests that the barrier domain of buccal membrane permeation is less lipophilic than octanol.
In the investigation of the relationship between permeant MW and the permeability of the polar pathway in the buccal mucosa using the macromolecules and pH independent permeants that have log Kow < −1, a correlation between log P and MW was observed. The result of the hindered transport analysis suggests an effective pore radius of 1.5 to 3 nm in the buccal mucosa for the permeation of polar permeants and macromolecules. This pore size value is in the same order of magnitude as other biological membranes such as skin, cornea, conjunctiva, nail, and GI mucosal monolayer.
It should be pointed out that the present review is focused on porcine buccal mucosa because (a) the majority of the permeability data in previous studies were from this animal model and (b) the similarity between human and porcine buccal mucosae. In addition, the permeability data presented in this review can be different from the permeability for drug delivery in practice because drug formulations usually contain excipients that can enhance membrane permeability for more effective drug delivery and this review is focused only on the intrinsic permeability of the buccal mucosa. The conclusion from the analyses (model of Equation (10)) using all the permeant data collected in this review is affected by (a) data variabilities in the previous studies (i.e., variabilities of P, Kow, and Dow), (b) model limitations such as the influence of the polar pathway and the difference between Km and Kow, and (c) uncertainties in permeant pH-ionization relationship (i.e., the use of Dow) arise from undefined membrane pH that can be different from the solution pH. The model of Equation (6) could account for the difference between Km and Kow but not the other factors. Hence, there was a need to apply certain exclusion criteria in the analyses, leading to a smaller dataset, in order to generate meaningful results. Although a more comprehensive analysis of buccal permeability could not be completed in the present review due to the availability and variability of the data in the literature, the results provide insights into possible quantitative structure permeability relationships of the buccal mucosa.

8. Conclusions

The buccal mucosa provides a number of advantages as an alternative route of drug administration. To develop more effective buccal drug delivery systems, knowledge on the intrinsic permeability of the buccal mucosa is essential. However, an extensive review of the permeability data is not available despite the numerous studies and reviews on the topic of buccal drug delivery. In the present review, the intrinsic permeability coefficients of porcine buccal mucosa were collected, a database of the permeability coefficients was generated, and the influences of permeant lipophilicity (as log Kow and log Dow) and molecular size (as MW) on the permeability of the buccal mucosa were analyzed. The first observation was the large variability among the published permeability data (of the same permeants). Such variability in buccal permeability studies in the literature could lead to poor correlation (coefficient of determination) in analyzing the permeability data for a possible quantitative structure permeability relationship. In the analyses of all permeability data including the permeants with pH dependent ionization, the permeability was described better by log Dow than log Kow for membrane partitioning and permeation, but the correlation was relatively poor with no observable effect of permeant MW. This can be attributed to the difference between solution and membrane pH when using the fraction of unionized permeant in the solution to account for membrane permeation. For the permeability data of pH independent permeants (which are not affected by ionization due to pH in the analysis), a better correlation was observed and the correlation improved with the incorporation of MW as the additional independent variable. The analysis of the relationship between the permeability and partition coefficient of the permeants for the buccal membrane barrier suggested an apparent linear free energy relationship that was less lipophilic than octanol. For the permeability data of macromolecules and polar permeants, an effective pore radius of 1.5 to 3 nm was found for the buccal mucosa using the hindered transport theory. The results obtained in this review could improve our understanding of the buccal mucosal barrier and assist in the development of more effective buccal drug delivery and dosage forms.

Author Contributions

Conceptualization, S.K.L.; Methodology, S.K.L.; Formal Analysis, A.W., S.K.L.; Investigation, A.W., H.A.C., P.B.P., S.K.L.; Resources, S.K.L.; Data Curation, A.W., H.A.C., P.B.P., S.K.L.; Writing—Original Draft Preparation, S.K.L.; Writing—Review & Editing, A.W., H.A.C., P.B.P., S.K.L.; Visualization, S.K.L.; Supervision, S.K.L.; Project Administration, S.K.L.; Funding Acquisition, S.K.L. All authors have read and agreed to the published version of the manuscript.

Funding

The research in this publication was supported in part by National Institute of Dental & Craniofacial Research (NIDCR) of the National Institutes of Health (NIH) under Award Number R15 DE028701. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank Michael Murawsky for his inputs.

Conflicts of Interest

The authors have no conflicts of interest.

Appendix A. Permeability Data of Porcine Buccal Mucosa

Table A1. Properties of permeants (MW, log Kow, and pKa), permeability coefficients, and sources of buccal permeation data.
Table A1. Properties of permeants (MW, log Kow, and pKa), permeability coefficients, and sources of buccal permeation data.
PermeantMWlog KowpKapHlog Dowlog P (cm/s)Ref
Acebutolol336.41.719.466.8−1.56−7.57[28]
Acebutolol336.41.719.467.4−0.23−7.40[34]
Acyclovir225.2−1.562.52, 9.356.8−1.56−5.21[35]
Acyclovir225.2−1.562.52, 9.353.3~8.8−1.56−5.20 d[36]
Alprenolol249.43.19.57.41.18−5.70[34]
Amitriptyline277.45.049.316.81.64−4.89[27]
Antipyrine188.20.391.456.80.39−5.65[37]
Antipyrine188.20.391.456.80.39−5.27[27]
Antipyrine188.20.381.46.80.38−5.67[29]
Atenolol266.30.229.547.4−0.80−7.30[34]
Atenolol266.30.229.547.4−1.92−7.39[38]
Atenolol266.30.229.546.8−1.30−7.55[27]
Atenolol266.30.229.546.8−1.30−6.88[28]
Atenolol266.30.229.547.25−2.07−6.72[39]
Atenolol a266.30.229.546.8--- c--- a[40]
Benznidazole a260.20.91--- c--- c0.91--- a[41]
Bupivacaine288.43.418.16.82.09−5.10[37]
Bupivacaine288.43.78.16.82.38−4.77[27]
Bupivacaine288.43.418.16.82.09−4.80[29]
Buspirone385.52.634.12, 7.32varying pH
unionized form data
2.63−5.54[42]
Buspirone385.52.634.12, 7.326.82.00−5.04[37]
Buspirone385.52.634.12, 7.326.82.80−4.81[27]
Buspirone385.52.634.12, 7.326.82.00−4.76[29]
Caffeine194.2−0.0710.46.8−0.07−5.05[27]
Caffeine194.2−0.0710.4--- c−0.07−4.70[43]
Caffeine194.2−0.0710.46.8−0.07−5.49[37]
Caffeine194.2−0.0710.4--- c−0.07−4.63 d[44]
Caffeine194.2−0.0710.46.8−0.07−5.09[29]
Caffeine194.2−0.0710.46.8−0.07−4.96[45]
Caffeine194.2−0.0710.47.25−0.07−5.57[39]
Carbamazepine236.32.4513.96.752.45−5.09[46]
Carprofen273.73.84.47.40.80−2.63[47]
Carvedilol a406.54.197.87.43.64--- a[48]
Carvedilol406.54.197.87.43.64−5.24[49]
Carvedilol a406.54.197.87.43.64--- a[50]
Cathine151.20.839.27.25−1.12−5.50[39]
Cathinone 149.21.389.27.25−0.57−5.52[39]
Decitabine228.2−1.89--- c7.0−1.89−7.64 d[51]
Diazepam284.72.823.46.82.82−5.37[52]
Diazepam284.72.823.46.82.82−5.32[45]
Didanosine236.2−0.7549.137.4−0.754−5.92 d[53]
Didanosine236.2−0.7549.137.4−0.754−4.50[54]
Didanosine236.2−0.7549.137.4−0.754−6.19[55]
Dideoxycytidine211.2−1.3--- c7.1−1.3−5.39[56]
Dideoxycytidine211.2−1.3--- c7.4−1.3−6.76[57]
Diltiazem414.52.797.76.81.04−5.14[27]
Diltiazem414.52.797.76.01.08−6.34[58]
Donepezil379.54.868.97.43.35−4.45[59]
Doxepin279.44.298.964.70.03−6.87[60]
Endomorphin-1610.7--- c--- c7.1--- c−6.25[61]
Estradiol272.44.01--- c7.44.01−4.29 d[44]
Estradiol272.44.01--- c7.44.01−4.54[43]
Estradiol a272.44.01--- c7.44.01--- a[62]
Felodipine384.23.865.077.43.86−5.06[63]
Fentanyl336.54.0597.01.41−5.15[64]
FITC (fluorescein isothiocyanate)389.4--- c--- c--- c--- c−7.52[65]
FITC-dextran4000--- c--- c--- c--- c−7.95[65]
FITC-dextran4400--- c--- c6.4--- c−7.83[66]
FITC-dextran10,000--- c--- c--- c--- c−7.53[65]
FITC-dextran20,000--- c--- c--- c--- c<−9.3 e[65]
FITC-dextran40,000--- c--- c--- c--- c<−9.3 e[65]
Flecainide414.33.789.37.40.05−7.19[67]
Furosemide330.72.034.76.8−1.00−8.48[27]
Galantamine287.351.87.976.80.60−4.93[68]
Hydrocortisone acetate a404.52.19--- c--- c2.19--- a[69]
Insulin5778--- c--- c7.4--- c−8.18[70]
Irinotecan CPT-11586.73.29.37.40.80−4.24[71]
Isoniazid137.1−0.71.86.8--- c−6.30[72]
Ketoprofen254.33.124.457.4--- c−5.45[73]
Labetalol328.43.099.37.40.52−7.70[34]
Labetalol328.43.099.36.8−0.03−6.26[28]
Lamotrigine265.10.895.77.40.89−5.19[74]
Lidocaine234.32.17.96.80.97−4.77[27]
Lidocaine234.32.447.96.00.53−6.30[58]
Lidocaine234.32.447.97.01.49−6.57[75]
Lidocaine a234.32.447.97.0--- c--- a[76]
Lidocaine234.32.17.97.40.97−5.73[73]
Lidocaine234.32.447.97.01.49−5.41[77]
Mannitol182.2−3.1--- c7.4−3.1−6.70[34]
Mannitol182.2−3.1--- c7.4−3.1−6.70[78]
Mannitol182.2−3.1--- c--- c−3.1−5.60[79]
Mannitol182.2−3.1--- c6.8−3.1−6.08 d[80]
Mannitol182.2−3.1--- c7.4−3.1−7.00[81]
Metoprolol267.41.959.567.4−0.07−5.52[34]
Metoprolol267.41.959.567.4−0.21−4.97[78]
Metoprolol267.41.959.566.8−0.81−5.89[27]
Metoprolol267.41.959.566.8−0.56−8.60[28]
Metoprolol267.41.959.567.4, 8.0, 8.5, 9.5 f−0.21−5.10, −4.46, −4.30, −4.14 f[82]
Morphine285.30.898.216.8−0.70−5.72 d[83]
Morphine285.30.898.21--- c−0.70−5.88[84]
Naltrexone341.41.928.16.80.60−4.76[85]
Naltrexone341.41.928.16.80.60−5.28[86]
Naproxen230.33.184.26.80.58−5.42[27]
Nicotine162.21.173.04, 7.84varying pH
unionized form data
1.17−5.00[87]
Nicotine162.21.173.4, 8.2unionized form data1.17−4.88[88]
Nicotine162.21.33.26, 8.067.40.55−7.81[89]
Nicotine162.21.173.04, 7.844.0−2.67−7.90[58]
Nicotine162.21.173.4, 8.26.8−0.25−5.16[52]
Nicotine162.21.173.4, 8.26.8−0.25−5.26 d[79]
Nicotine162.21.173.4, 8.26.8−0.25−5.10[90]
Nicotine162.21.173.4, 8.26.8−0.25−4.50 d[80]
Nimesulide308.31.946.56.81.46−4.52[27]
Norephedrine151.20.679.447.25−1.52−5.67[39]
Oligonucleotide6405--- c--- c--- c--- c−8.96[91]
Omeprazole345.42.234.87.02.23−5.73[92]
Ondansetron293.42.47.34unionized form data2.4−5.31[93]
Ondansetron293.42.47.344.0−0.94−6.75[94]
Oxprenolol265.42.19.677.40.88−5.52[34]
Oxprenolol265.42.19.676.80.81−6.05[28]
Pindolol248.31.839.547.4−0.31−6.70[34]
Pindolol248.31.839.546.8−0.91−6.92[27]
Pioglitazone356.40.4255.197.40.42−5.18[63]
Pioglitazone356.40.4255.19--- c0.42−6.15[95]
Polyethylene glycol282−2--- c7.4−2.0−5.53[96]
Polyethylene glycol326−2.1--- c7.4−2.1−5.57[96]
Polyethylene glycol370−2.15--- c7.4−2.15−5.58[96]
Polyethylene glycol414−2.2--- c7.4−2.2−5.60[96]
Polyethylene glycol458−2.2--- c7.4−2.2−5.66[96]
Polyethylene glycol502−2.25--- c7.4−2.25−5.80[96]
Polyethylene glycol546−2.3--- c7.4−2.3−5.89[96]
Prilocaine a220.32.117.897.4--- c--- a[76]
Propranolol259.33.489.457.41.28−5.89[34]
Propranolol259.33.489.456.80.83−4.85[27]
Propranolol259.33.489.456.81.20−5.80[28]
Propranolol259.33.489.457.41.43−5.64[97]
Salbutamol a239.31.410.37.1−1.80--- a[98]
Salmon calcitonin3431.9--- c--- c4.0--- c−8.60[99]
Salmon calcitonin3431.9--- c--- c7.4--- c−8.86[100]
Saquinavir670.84.77.17.44.52−4.26[101]
Sotalol272.40.249.8, 8.37.4−2.16−7.02[67]
Steroidal glycoside P57 (extract) b--- c--- c--- c7.0--- c−4.66 b[102]
Tacrine a198.32.719.957.40.31--- a[103]
Tenofovir287.2−1.6--- c--- c−1.6−4.95[54]
Tertatolol295.42.099.87.4−0.37−6.00[34]
Testosterone288.43.32--- c7.43.32−5.96[34]
Tetramethylpyrazine136.21.28--- c--- c1.28−5.04[104]
Thiocolchicoside563.60.34--- c6.80.34−6.72[105]
Timolol316.41.839.217.4−1.71−6.52[34]
Triamcinolone acetonide434.52.53--- c7.42.53−5.39[106]
Triamcinolone acetonide434.52.53--- c--- c2.53−5.55[43]
Triamcinolone acetonide434.52.53--- c--- c2.53−5.60[107]
Triamcinolone acetonide434.52.53--- c6.752.53−5.52[46]
Verapamil454.63.798.96.81.69−4.60[27]
Warfarin308.32.64.96.80.69−5.80[27]
Water18−1.38--- c7.4−1.38−5.09[34]
Water18−1.38--- c7.4−1.38−4.94[81]
a Permeability data with formulations involving organic solvents or enhancers in the permeation experiments. The data were not used in the present analysis. b Not sufficient information for use in the present analysis. c Not applicable or not available. d Multiple permeability values are available in the reference and the average value of log p was used. e Below detection limit. f Multiple pH conditions are available.

Appendix B. Derivation of Equations in Model Analyses

From Equation (2) in “Membrane Transport Theory” using a parallel pathway permeation model of lipoidal and polar pathways [120,121], the permeability coefficient of the membrane (P) can be expressed as [32]:
P = P l f u + P p
where fu is the fraction of unionized permeant and subscripts l and p represent the lipoidal and polar pathways, respectively. The fraction of unionized permeant is related to the octanol/water distribution coefficient (Dow) and octanol/water partition coefficient (Kow).
D o w = K o w f u
where   f u = 1 / 1 + 10 p H p K a   for   weak   acid
and   f u = 1 / 1 + 10 p K a p H   for   weak   base
Combining Equations (A1) and (A2),
P = P l D o w / K o w + P p
Assuming that the microenvironment of the lipid lamellae in the membrane can be mimicked by octanol, membrane partition coefficient can be estimated by Kow (i.e., Km = Kow):
P l = K m D m / h m = K o w D m / h m
where Dm is membrane diffusion coefficient, Km is membrane partition coefficient, and hm is effective thickness of the membrane. The relationship between the permeability of lipid membrane (e.g., lipid bilayer) and permeant lipophilicity defined by its Kow in Equation (A6) is commonly used in membrane permeation model. Combining Equations (A5) and (A6),
P = D o w D m / h m + P p
When the contribution of Pp is minimal, Equation (A7) can be rewritten as:
log P = log D o w + log D m + h m
where hm is a constant related to the effective thickness. The membrane diffusion coefficient can be expressed as [31]:
log D m = log D 0 + c   M W
where MW is permeant molecular weight, D0 is the hypothetical diffusion coefficient of permeants with zero molecular volume, and c is a constant. Equation (A8) can be rewritten as:
log P = log D o w + c   M W + c o n s t a n t
where constant is a constant in the relationship. Or,
log P = log K o w + c   M W + c o n s t a n t
when Dow = Kow for permeants that are not affected by pH.
A linear free energy relationship can be used to describe the microenvironment of the lipid barrier when it is different from that of octanol for membrane partitioning [122]:
K m = b K o w a
Replacing Kow in Equation A6 by the relationship of Equation (A12) and rearranging the equation as in the derivation from Equations (A5)–(A10),
log ( P / D o w ) = a 1 log K o w + c   M W + c o n s t a n t

References

  1. Hao, J.; Heng, P.W. Buccal delivery systems. Drug Dev. Ind. Pharm. 2003, 29, 821–832. [Google Scholar] [CrossRef] [PubMed]
  2. Birudaraj, R.; Mahalingam, R.; Li, X.; Jasti, B.R. Advances in buccal drug delivery. Crit. Rev. Ther. Drug Carr. Syst. 2005, 22, 295–330. [Google Scholar] [CrossRef]
  3. Pinto, S.; Pintado, M.E.; Sarmento, B. In vivo, ex vivo and in vitro assessment of buccal permeation of drugs from delivery systems. Expert Opin. Drug Deliv. 2020, 17, 33–48. [Google Scholar] [CrossRef]
  4. Patel, V.F.; Liu, F.; Brown, M.B. Modeling the oral cavity: In vitro and in vivo evaluations of buccal drug delivery systems. J. Control. Release 2012, 161, 746–756. [Google Scholar] [CrossRef]
  5. Harris, D.; Robinson, J.R. Drug delivery via the mucous membranes of the oral cavity. J. Pharm. Sci. 1992, 81, 1–10. [Google Scholar] [CrossRef]
  6. Shojaei, A.H. Buccal mucosa as a route for systemic drug delivery: A review. J. Pharm. Pharm. Sci. 1998, 1, 15–30. [Google Scholar]
  7. Patel, V.F.; Liu, F.; Brown, M.B. Advances in oral transmucosal drug delivery. J. Control. Release 2011, 153, 106–116. [Google Scholar] [CrossRef] [Green Version]
  8. Senel, S.; Rathbone, M.J.; Cansiz, M.; Pather, I. Recent developments in buccal and sublingual delivery systems. Expert Opin. Drug Deliv. 2012, 9, 615–628. [Google Scholar] [CrossRef] [PubMed]
  9. Sattar, M.; Sayed, O.M.; Lane, M.E. Oral transmucosal drug delivery--current status and future prospects. Int. J. Pharm. 2014, 471, 498–506. [Google Scholar] [CrossRef] [PubMed]
  10. Nicolazzo, J.A.; Reed, B.L.; Finnin, B.C. Buccal penetration enhancers—How do they really work? J. Control. Release 2005, 105, 1–15. [Google Scholar] [CrossRef] [PubMed]
  11. Hassan, N.; Ahad, A.; Ali, M.; Ali, J. Chemical permeation enhancers for transbuccal drug delivery. Expert Opin. Drug Deliv. 2010, 7, 97–112. [Google Scholar] [CrossRef] [PubMed]
  12. Laffleur, F. Mucoadhesive polymers for buccal drug delivery. Drug Dev. Ind. Pharm. 2014, 40, 591–598. [Google Scholar] [CrossRef] [PubMed]
  13. Tran, P.H.L.; Duan, W.; Tran, T.T.D. Recent developments of nanoparticle-delivered dosage forms for buccal delivery. Int. J. Pharm. 2019, 571, 118697. [Google Scholar] [CrossRef] [PubMed]
  14. Macedo, A.S.; Castro, P.M.; Roque, L.; Thome, N.G.; Reis, C.P.; Pintado, M.E.; Fonte, P. Novel and revisited approaches in nanoparticle systems for buccal drug delivery. J. Control. Release 2020, 320, 125–141. [Google Scholar] [CrossRef] [PubMed]
  15. Singla, A.K.; Chawla, M.; Singh, A. Potential applications of carbomer in oral mucoadhesive controlled drug delivery system: A review. Drug Dev. Ind. Pharm. 2000, 26, 913–924. [Google Scholar] [CrossRef]
  16. Salamat-Miller, N.; Chittchang, M.; Johnston, T.P. The use of mucoadhesive polymers in buccal drug delivery. Adv. Drug Deliv. Rev. 2005, 57, 1666–1691. [Google Scholar] [CrossRef]
  17. Sudhakar, Y.; Kuotsu, K.; Bandyopadhyay, A.K. Buccal bioadhesive drug delivery--a promising option for orally less efficient drugs. J. Control. Release 2006, 114, 15–40. [Google Scholar] [CrossRef]
  18. Wanasathop, A.; Li, S.K. Iontophoretic drug delivery in the oral cavity. Pharmaceutics 2018, 10, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Mystakidou, K.; Katsouda, E.; Parpa, E.; Vlahos, L.; Tsiatas, M.L. Oral transmucosal fentanyl citrate: Overview of pharmacological and clinical characteristics. Drug Deliv. 2006, 13, 269–276. [Google Scholar] [CrossRef] [PubMed]
  20. Dinsmore, W.W.; Wyllie, M.G. The long-term efficacy and safety of a testosterone mucoadhesive buccal tablet in testosterone-deficient men. BJU Int. 2012, 110, 162–169. [Google Scholar] [CrossRef]
  21. Garnock-Jones, K.P. Fentanyl buccal soluble film: A review in breakthrough cancer pain. Clin. Drug Investig. 2016, 36, 413–419. [Google Scholar] [CrossRef] [PubMed]
  22. Pergolizzi, J.V., Jr.; Raffa, R.B.; Fleischer, C.; Zampogna, G.; Taylor, R., Jr. Management of moderate to severe chronic low back pain with buprenorphine buccal film using novel bioerodible mucoadhesive technology. J. Pain Res. 2016, 9, 909–916. [Google Scholar] [CrossRef] [Green Version]
  23. Smart, J.D. Buccal drug delivery. Expert Opin. Drug Deliv. 2005, 2, 507–517. [Google Scholar] [CrossRef]
  24. Kraan, H.; Vrieling, H.; Czerkinsky, C.; Jiskoot, W.; Kersten, G.; Amorij, J.P. Buccal and sublingual vaccine delivery. J. Control. Release 2014, 190, 580–592. [Google Scholar] [CrossRef] [Green Version]
  25. Morales, J.O.; McConville, J.T. Novel strategies for the buccal delivery of macromolecules. Drug Dev. Ind. Pharm. 2014, 40, 579–590. [Google Scholar] [CrossRef] [PubMed]
  26. Caon, T.; Jin, L.; Simoes, C.M.; Norton, R.S.; Nicolazzo, J.A. Enhancing the buccal mucosal delivery of peptide and protein therapeutics. Pharm. Res. 2015, 32, 1–21. [Google Scholar] [CrossRef] [PubMed]
  27. Kokate, A.; Li, X.; Williams, P.J.; Singh, P.; Jasti, B.R. In silico prediction of drug permeability across buccal mucosa. Pharm. Res. 2009, 26, 1130–1139. [Google Scholar] [CrossRef]
  28. Amores, S.; Lauroba, J.; Calpena, A.; Colom, H.; Gimeno, A.; Domenech, J. A comparative ex vivo drug permeation study of beta-blockers through porcine buccal mucosa. Int. J. Pharm. 2014, 468, 50–54. [Google Scholar] [CrossRef]
  29. Kulkarni, U.D.; Mahalingam, R.; Li, X.; Pather, I.; Jasti, B. Effect of experimental temperature on the permeation of model diffusants across porcine buccal mucosa. AAPS PharmSciTech 2011, 12, 579–586. [Google Scholar] [CrossRef] [Green Version]
  30. Gandhi, R.B.; Robinson, J.R. Oral cavity as a site for bioadhesive drug delivery. Adv. Drug Deliv. Rev. 1994, 13, 43–74. [Google Scholar] [CrossRef]
  31. Potts, R.O.; Guy, R.H. Predicting skin permeability. Pharm. Res. 1992, 9, 663–669. [Google Scholar] [CrossRef] [PubMed]
  32. Adson, A.; Burton, P.S.; Raub, T.J.; Barsuhn, C.L.; Audus, K.L.; Ho, N.F. Passive diffusion of weak organic electrolytes across Caco-2 cell monolayers: Uncoupling the contributions of hydrodynamic, transcellular, and paracellular barriers. J. Pharm. Sci. 1995, 84, 1197–1204. [Google Scholar] [CrossRef]
  33. Tsutsumi, K.; Li, S.K.; Ghanem, A.H.; Ho, N.F.; Higuchi, W.I. A systematic examination of the in vitro Ussing chamber and the in situ single-pass perfusion model systems in rat ileum permeation of model solutes. J. Pharm. Sci. 2003, 92, 344–359. [Google Scholar] [CrossRef] [PubMed]
  34. Nielsen, H.M.; Rassing, M.R. TR146 cells grown on filters as a model of human buccal epithelium: IV. Permeability of water, mannitol, testosterone and beta-adrenoceptor antagonists. Comparison to human, monkey and porcine buccal mucosa. Int. J. Pharm. 2000, 194, 155–167. [Google Scholar] [CrossRef]
  35. Shojaei, A.H.; Berner, B.; Xiaoling, L. Transbuccal delivery of acyclovir: I. In vitro determination of routes of buccal transport. Pharm. Res. 1998, 15, 1182–1188. [Google Scholar] [CrossRef] [PubMed]
  36. Shojaei, A.H.; Zhuo, S.L.; Li, X. Transbuccal delivery of acyclovir (II): Feasibility, system design, and in vitro permeation studies. J. Pharm. Pharm. Sci. 1998, 1, 66–73. [Google Scholar]
  37. Kulkarni, U.; Mahalingam, R.; Pather, S.I.; Li, X.; Jasti, B. Porcine buccal mucosa as an in vitro model: Relative contribution of epithelium and connective tissue as permeability barriers. J. Pharm. Sci. 2009, 98, 471–483. [Google Scholar] [CrossRef] [PubMed]
  38. Jacobsen, J. Buccal iontophoretic delivery of atenolol·HCl employing a new in vitro three-chamber permeation cell. J. Control. Release 2001, 70, 83–95. [Google Scholar] [CrossRef]
  39. Atlabachew, M.; Combrinck, S.; Viljoen, A.M.; Hamman, J.H.; Gouws, C. Isolation and in vitro permeation of phenylpropylamino alkaloids from Khat (Catha edulis) across oral and intestinal mucosal tissues. J. Ethnopharmacol. 2016, 194, 307–315. [Google Scholar] [CrossRef] [PubMed]
  40. Adhikari, S.N.R.; Panda, S. Buccal patches of atenolol formulated using fenugreek (Trigonella foenum-graecum L.) seed mucilage. Polim. Med. 2017, 47, 5–11. [Google Scholar] [CrossRef] [Green Version]
  41. Amaral, B.R.; Argenta, D.F.; Kroth, R.; Caon, T. Transbuccal delivery of benznidazole associated with monoterpenes: Permeation studies and mechanistic insights. Eur. J. Pharm. Sci. 2020, 143, 105177. [Google Scholar] [CrossRef] [PubMed]
  42. Birudaraj, R.; Berner, B.; Shen, S.; Li, X. Buccal permeation of buspirone: Mechanistic studies on transport pathways. J. Pharm. Sci. 2005, 94, 70–78. [Google Scholar] [CrossRef] [PubMed]
  43. Nicolazzo, J.A.; Reed, B.L.; Finnin, B.C. Modification of buccal drug delivery following pretreatment with skin penetration enhancers. J. Pharm. Sci. 2004, 93, 2054–2063. [Google Scholar] [CrossRef]
  44. Nicolazzo, J.A.; Reed, B.L.; Finnin, B.C. The effect of various in vitro conditions on the permeability characteristics of the buccal mucosa. J. Pharm. Sci. 2003, 92, 2399–2410. [Google Scholar] [CrossRef] [PubMed]
  45. Hansen, S.E.; Marxen, E.; Janfelt, C.; Jacobsen, J. Buccal delivery of small molecules—Impact of levulinic acid, oleic acid, sodium dodecyl sulfate and hypotonicity on ex vivo permeability and spatial distribution in mucosa. Eur. J. Pharm. Biopharm. 2018, 133, 250–257. [Google Scholar] [CrossRef] [PubMed]
  46. Caon, T.; Simoes, C.M. Effect of freezing and type of mucosa on ex vivo drug permeability parameters. AAPS PharmSciTech 2011, 12, 587–592. [Google Scholar] [CrossRef] [Green Version]
  47. Gomez-Segura, L.; Parra, A.; Calpena, A.C.; Gimeno, A.; Boix-Montanes, A. Carprofen permeation test through porcine ex vivo mucous membranes and ophthalmic tissues for tolerability assessments: Validation and histological study. Vet. Sci. 2020, 7, 152. [Google Scholar] [CrossRef]
  48. Yamsani, V.V.; Gannu, R.; Kolli, C.; Rao, M.E.; Yamsani, M.R. Development and in vitro evaluation of buccoadhesive carvedilol tablets. Acta Pharm. 2007, 57, 185–197. [Google Scholar] [CrossRef] [Green Version]
  49. Chen, J.; Duan, H.; Pan, H.; Yang, X.; Pan, W. Two types of core/shell fibers based on carboxymethyl chitosan and Sodium carboxymethyl cellulose with self-assembled liposome for buccal delivery of carvedilol across TR146 cell culture and porcine buccal mucosa. Int. J. Biol. Macromol. 2019, 128, 700–709. [Google Scholar] [CrossRef]
  50. Vishnu, Y.V.; Chandrasekhar, K.; Ramesh, G.; Rao, Y.M. Development of mucoadhesive patches for buccal administration of carvedilol. Curr. Drug Deliv. 2007, 4, 27–39. [Google Scholar] [CrossRef]
  51. Mahalingam, R.; Ravivarapu, H.; Redkar, S.; Li, X.; Jasti, B.R. Transbuccal delivery of 5-aza-2’-deoxycytidine: Effects of drug concentration, buffer solution, and bile salts on permeation. AAPS PharmSciTech 2007, 8, E28–E33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Marxen, E.; Axelsen, M.C.; Pedersen, A.M.L.; Jacobsen, J. Effect of cryoprotectants for maintaining drug permeability barriers in porcine buccal mucosa. Int. J. Pharm. 2016, 511, 599–605. [Google Scholar] [CrossRef]
  53. Ojewole, E.; Mackraj, I.; Akhundov, K.; Hamman, J.; Viljoen, A.; Olivier, E.; Wesley-Smith, J.; Govender, T. Investigating the effect of Aloe vera gel on the buccal permeability of didanosine. Planta Med. 2012, 78, 354–361. [Google Scholar] [CrossRef] [Green Version]
  54. Rambharose, S.; Ojewole, E.; Mackraj, I.; Govender, T. Comparative buccal permeability enhancement of didanosine and tenofovir by potential multifunctional polymeric excipients and their effects on porcine buccal histology. Pharm. Dev. Technol. 2014, 19, 82–90. [Google Scholar] [CrossRef]
  55. Ojewole, E.; Kalhapure, R.; Akamanchi, K.; Govender, T. Novel oleic acid derivatives enhance buccal permeation of didanosine. Drug Dev. Ind. Pharm. 2014, 40, 657–668. [Google Scholar] [CrossRef] [PubMed]
  56. Shojaei, A.H.; Khan, M.; Lim, G.; Khosravan, R. Transbuccal permeation of a nucleoside analog, dideoxycytidine: Effects of menthol as a permeation enhancer. Int. J. Pharm. 1999, 192, 139–146. [Google Scholar] [CrossRef]
  57. Xiang, J.; Fang, X.; Li, X. Transbuccal delivery of 2′,3′-dideoxycytidine: In vitro permeation study and histological investigation. Int. J. Pharm. 2002, 231, 57–66. [Google Scholar] [CrossRef]
  58. Hu, L.; Silva, S.M.; Damaj, B.B.; Martin, R.; Michniak-Kohn, B.B. Transdermal and transbuccal drug delivery systems: Enhancement using iontophoretic and chemical approaches. Int. J. Pharm. 2011, 421, 53–62. [Google Scholar] [CrossRef]
  59. Caon, T.; Pan, Y.; Simoes, C.M.; Nicolazzo, J.A. Exploiting the buccal mucosa as an alternative route for the delivery of donepezil hydrochloride. J. Pharm. Sci. 2014, 103, 1643–1651. [Google Scholar] [CrossRef]
  60. Gimeno, A.; Calpena, A.C.; Sanz, R.; Mallandrich, M.; Peraire, C.; Clares, B. Transbuccal delivery of doxepin: Studies on permeation and histological investigation. Int. J. Pharm. 2014, 477, 650–654. [Google Scholar] [CrossRef]
  61. Bird, A.P.; Faltinek, J.R.; Shojaei, A.H. Transbuccal peptide delivery: Stability and in vitro permeation studies on endomorphin-1. J. Control. Release 2001, 73, 31–36. [Google Scholar] [CrossRef]
  62. Nicolazzo, J.A.; Reed, B.L.; Finnin, B.C. Enhanced buccal mucosal retention and reduced buccal permeability of estradiol in the presence of padimate O and Azone: A mechanistic study. J. Pharm. Sci. 2005, 94, 873–882. [Google Scholar] [CrossRef] [PubMed]
  63. Palem, C.R.; Gannu, R.; Yamsani, S.K.; Yamsani, V.V.; Yamsani, M.R. Development of bioadhesive buccal tablets for felodipine and pioglitazone in combined dosage form: In vitro, ex vivo, and in vivo characterization. Drug Deliv. 2011, 18, 344–352. [Google Scholar] [CrossRef] [PubMed]
  64. Diaz Del Consuelo, I.; Pizzolato, G.P.; Falson, F.; Guy, R.H.; Jacques, Y. Evaluation of pig esophageal mucosa as a permeability barrier model for buccal tissue. J. Pharm. Sci. 2005, 94, 2777–2788. [Google Scholar] [CrossRef] [PubMed]
  65. Hoogstraate, A.J.; Cullander, C.; Nagelkerke, J.F.; Senel, S.; Verhoef, J.C.; Junginger, H.E.; Bodde, H.E. Diffusion rates and transport pathways of fluorescein isothiocyanate (FITC)-labeled model compounds through buccal epithelium. Pharm. Res. 1994, 11, 83–89. [Google Scholar] [CrossRef] [PubMed]
  66. Sandri, G.; Rossi, S.; Bonferoni, M.C.; Ferrari, F.; Zambito, Y.; Di Colo, G.; Caramella, C. Buccal penetration enhancement properties of N-trimethyl chitosan: Influence of quaternization degree on absorption of a high molecular weight molecule. Int. J. Pharm. 2005, 297, 146–155. [Google Scholar] [CrossRef]
  67. Deneer, V.H.; Drese, G.B.; Roemele, P.E.; Verhoef, J.C.; Lie, A.H.L.; Kingma, J.H.; Brouwers, J.R.; Junginger, H.E. Buccal transport of flecainide and sotalol: Effect of a bile salt and ionization state. Int. J. Pharm. 2002, 241, 127–134. [Google Scholar] [CrossRef]
  68. De Caro, V.; Giandalia, G.; Siragusa, M.G.; Paderni, C.; Campisi, G.; Giannola, L.I. Evaluation of galantamine transbuccal absorption by reconstituted human oral epithelium and porcine tissue as buccal mucosa models: Part I. Eur. J. Pharm. Biopharm. 2008, 70, 869–873. [Google Scholar] [CrossRef] [PubMed]
  69. Ceschel, G.C.; Maffei, P.; Lombardi Borgia, S.; Ronchi, C. Design and evaluation of buccal adhesive hydrocortisone acetate (HCA) tablets. Drug Deliv. 2001, 8, 161–171. [Google Scholar] [CrossRef] [PubMed]
  70. Bashyal, S.; Seo, J.E.; Keum, T.; Noh, G.; Lamichhane, S.; Kim, J.H.; Kim, C.H.; Choi, Y.W.; Lee, S. Facilitated buccal insulin delivery via hydrophobic ion-pairing approach: In vitro and ex vivo evaluation. Int. J. Nanomed. 2021, 16, 4677–4691. [Google Scholar] [CrossRef]
  71. Shah, V.; Bellantone, R.A.; Taft, D.R. Evaluating the potential for delivery of irinotecan via the buccal route: Physicochemical characterization and in vitro permeation assessment across porcine buccal mucosa. AAPS PharmSciTech 2017, 18, 867–874. [Google Scholar] [CrossRef]
  72. Kroth, R.; Argenta, D.F.; Conte, J.; Amaral, B.R.; Caon, T. Transbuccal delivery of isoniazid: Ex vivo permeability and drug-surfactant interaction studies. AAPS PharmSciTech 2020, 21, 289. [Google Scholar] [CrossRef] [PubMed]
  73. Eleftheriadis, G.K.; Monou, P.K.; Bouropoulos, N.; Boetker, J.; Rantanen, J.; Jacobsen, J.; Vizirianakis, I.S.; Fatouros, D.G. Fabrication of mucoadhesive buccal films for local administration of ketoprofen and lidocaine hydrochloride by combining fused deposition modeling and inkjet printing. J. Pharm. Sci. 2020, 109, 2757–2766. [Google Scholar] [CrossRef] [PubMed]
  74. Mashru, R.; Sutariya, V.; Sankalia, M.; Sankalia, J. Transbuccal delivery of lamotrigine across porcine buccal mucosa: In vitro determination of routes of buccal transport. J. Pharm. Pharm. Sci. 2005, 8, 54–62. [Google Scholar]
  75. Franz-Montan, M.; Serpe, L.; Martinelli, C.C.; da Silva, C.B.; Santos, C.P.; Novaes, P.D.; Volpato, M.C.; de Paula, E.; Lopez, R.F.; Groppo, F.C. Evaluation of different pig oral mucosa sites as permeability barrier models for drug permeation studies. Eur. J. Pharm. Sci. 2016, 81, 52–59. [Google Scholar] [CrossRef] [PubMed]
  76. do Couto, R.O.; Cubayachi, C.; Calefi, P.L.; Lopez, R.F.V.; Pedrazzi, V.; De Gaitani, C.M.; de Freitas, O. Combining amino amide salts in mucoadhesive films enhances needle-free buccal anesthesia in adults. J. Control. Release 2017, 266, 205–215. [Google Scholar] [CrossRef]
  77. Abu-Huwaij, R.; Assaf, S.; Salem, M.; Sallam, A. Potential mucoadhesive dosage form of lidocaine hydrochloride: II. In vitro and in vivo evaluation. Drug Dev. Ind. Pharm. 2007, 33, 437–448. [Google Scholar] [CrossRef] [PubMed]
  78. Meng-Lund, E.; Marxen, E.; Pedersen, A.M.L.; Mullertz, A.; Hyrup, B.; Holm, R.; Jacobsen, J. Ex vivo correlation of the permeability of metoprolol across human and porcine buccal mucosa. J. Pharm. Sci. 2014, 103, 2053–2061. [Google Scholar] [CrossRef]
  79. Marxen, E.; Mosgaard, M.D.; Pedersen, A.M.L.; Jacobsen, J. Mucin dispersions as a model for the oromucosal mucus layer in in vitro and ex vivo buccal permeability studies of small molecules. Eur. J. Pharm. Biopharm. 2017, 121, 121–128. [Google Scholar] [CrossRef] [Green Version]
  80. Marxen, E.; Jin, L.; Jacobsen, J.; Janfelt, C.; Hyrup, B.; Nicolazzo, J.A. Effect of permeation enhancers on the buccal permeability of nicotine: Ex vivo transport studies complemented by MALDI MS imaging. Pharm. Res. 2018, 35, 70. [Google Scholar] [CrossRef] [PubMed]
  81. Lee, J.; Lee, S.K.; Choi, Y.W. The effect of storage conditions on the permeability of porcine buccal mucosa. Arch. Pharm. Res. 2002, 25, 546–549. [Google Scholar] [CrossRef] [PubMed]
  82. Holm, R.; Meng-Lund, E.; Andersen, M.B.; Jespersen, M.L.; Karlsson, J.J.; Garmer, M.; Jorgensen, E.B.; Jacobsen, J. In vitro, ex vivo and in vivo examination of buccal absorption of metoprolol with varying pH in TR146 cell culture, porcine buccal mucosa and Gottingen minipigs. Eur. J. Pharm. Sci. 2013, 49, 117–124. [Google Scholar] [CrossRef] [PubMed]
  83. Senel, S.; Duchene, D.; Hincal, A.A.; Capan, Y.; Ponchel, G. In vitro studies on enhancing effect of sodium glycocholate on transbuccal permeation of morphine hydrochloride. J. Control. Release 1998, 51, 107–113. [Google Scholar] [CrossRef] [PubMed]
  84. Senel, S.; Hincal, A.A. Drug permeation enhancement via buccal route: Possibilities and limitations. J. Control. Release 2001, 72, 133–144. [Google Scholar] [CrossRef]
  85. Giannola, L.I.; De Caro, V.; Giandalia, G.; Siragusa, M.G.; Tripodo, C.; Florena, A.M.; Campisi, G. Release of naltrexone on buccal mucosa: Permeation studies, histological aspects and matrix system design. Eur. J. Pharm. Biopharm. 2007, 67, 425–433. [Google Scholar] [CrossRef]
  86. Rai, V.; Tan, H.S.; Michniak-Kohn, B. Effect of surfactants and pH on naltrexone (NTX) permeation across buccal mucosa. Int. J. Pharm. 2011, 411, 92–97. [Google Scholar] [CrossRef] [Green Version]
  87. Nair, M.K.; Chetty, D.J.; Ho, H.; Chien, Y.W. Biomembrane permeation of nicotine: Mechanistic studies with porcine mucosae and skin. J. Pharm. Sci. 1997, 86, 257–262. [Google Scholar] [CrossRef] [PubMed]
  88. Chen, L.L.; Chetty, D.J.; Chien, Y.W. A mechanistic analysis to characterize oramucosal permeation properties. Int. J. Pharm. 1999, 184, 63–72. [Google Scholar] [CrossRef]
  89. Nielsen, H.M.; Rassing, M.R. Nicotine permeability across the buccal TR146 cell culture model and porcine buccal mucosa in vitro: Effect of pH and concentration. Eur. J. Pharm. Sci. 2002, 16, 151–157. [Google Scholar] [CrossRef]
  90. Marxen, E.; Jacobsen, J.; Hyrup, B.; Janfelt, C. Permeability barriers for nicotine and mannitol in porcine buccal mucosa studied by high-resolution MALDI mass spectrometry imaging. Mol. Pharm. 2018, 15, 519–526. [Google Scholar] [CrossRef] [PubMed]
  91. Jasti, B.R.; Zhou, S.; Mehta, R.C.; Li, X. Permeability of antisense oligonucleotide through porcine buccal mucosa. Int. J. Pharm. 2000, 208, 35–39. [Google Scholar] [CrossRef]
  92. Figueiras, A.; Hombach, J.; Veiga, F.; Bernkop-Schnurch, A. In vitro evaluation of natural and methylated cyclodextrins as buccal permeation enhancing system for omeprazole delivery. Eur. J. Pharm. Biopharm. 2009, 71, 339–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Mashru, R.C.; Sutariya, V.B.; Sankalia, M.G.; Sankalia, J.M. Effect of pH on in vitro permeation of ondansetron hydrochloride across porcine buccal mucosa. Pharm. Dev. Technol. 2005, 10, 241–247. [Google Scholar] [CrossRef] [PubMed]
  94. Hu, L.; Damaj, B.B.; Martin, R.; Michniak-Kohn, B.B. Enhanced in vitro transbuccal drug delivery of ondansetron HCl. Int. J. Pharm. 2011, 404, 66–74. [Google Scholar] [CrossRef]
  95. Silva-Abreu, M.; Espinoza, L.C.; Halbaut, L.; Espina, M.; Garcia, M.L.; Calpena, A.C. Comparative study of ex vivo transmucosal permeation of pioglitazone nanoparticles for the treatment of Alzheimer’s disease. Polymers 2018, 10, 316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Goswami, T.; Jasti, B.R.; Li, X. Estimation of the theoretical pore sizes of the porcine oral mucosa for permeation of hydrophilic permeants. Arch. Oral Biol. 2009, 54, 577–582. [Google Scholar] [CrossRef] [PubMed]
  97. Amores, S.; Domenech, J.; Colom, H.; Calpena, A.C.; Clares, B.; Gimeno, A.; Lauroba, J. An improved cryopreservation method for porcine buccal mucosa in ex vivo drug permeation studies using Franz diffusion cells. Eur. J. Pharm. Sci. 2014, 60, 49–54. [Google Scholar] [CrossRef]
  98. Prasanth, V.V.; Puratchikody, A.; Mathew, S.T.; Ashok, K.B. Effect of permeation enhancers in the mucoadhesive buccal patches of salbutamol sulphate for unidirectional buccal drug delivery. Res. Pharm. Sci. 2014, 9, 259–268. [Google Scholar]
  99. Oh, D.H.; Chun, K.H.; Jeon, S.O.; Kang, J.W.; Lee, S. Enhanced transbuccal salmon calcitonin (sCT) delivery: Effect of chemical enhancers and electrical assistance on in vitro sCT buccal permeation. Eur. J. Pharm. Biopharm. 2011, 79, 357–363. [Google Scholar] [CrossRef]
  100. Keum, T.; Noh, G.; Seo, J.E.; Bashyal, S.; Lee, S. In vitro and ex vivo evaluation of penetratin as a non-invasive permeation enhancer in the penetration of salmon calcitonin through TR146 buccal cells and porcine buccal tissues. Pharmaceuticals 2020, 13, 408. [Google Scholar] [CrossRef]
  101. Rambharose, S.; Ojewole, E.; Branham, M.; Kalhapure, R.; Govender, T. High-energy ball milling of saquinavir increases permeability across the buccal mucosa. Drug Dev. Ind. Pharm. 2014, 40, 639–648. [Google Scholar] [CrossRef]
  102. Vermaak, I.; Viljoen, A.M.; Chen, W.; Hamman, J.H. In vitro transport of the steroidal glycoside P57 from Hoodia gordonii across excised porcine intestinal and buccal tissue. Phytomedicine 2011, 18, 783–787. [Google Scholar] [CrossRef]
  103. Gore, A.V.; Liang, A.C.; Chien, Y.W. Comparative biomembrane permeation of tacrine using Yucatan minipigs and domestic pigs as the animal model. J. Pharm. Sci. 1998, 87, 441–447. [Google Scholar] [CrossRef] [PubMed]
  104. Liu, C.; Xu, H.N.; Li, X.L. In vitro permeation of tetramethylpyrazine across porcine buccal mucosa. Acta Pharmacol. Sin. 2002, 23, 792–796. [Google Scholar] [PubMed]
  105. Artusi, M.; Santi, P.; Colombo, P.; Junginger, H.E. Buccal delivery of thiocolchicoside: In vitro and in vivo permeation studies. Int. J. Pharm. 2003, 250, 203–213. [Google Scholar] [CrossRef]
  106. Shin, S.C.; Kim, J.Y. Enhanced permeation of triamcinolone acetonide through the buccal mucosa. Eur. J. Pharm. Biopharm. 2000, 50, 217–220. [Google Scholar] [CrossRef]
  107. Nicolazzo, J.A.; Reed, B.L.; Finnin, B.C. Enhancing the buccal mucosal uptake and retention of triamcinolone acetonide. J. Control. Release 2005, 105, 240–248. [Google Scholar] [CrossRef] [PubMed]
  108. NIH. PubChem. Available online: https://pubchem.ncbi.nlm.nih.gov/ (accessed on 19 September 2021).
  109. Tsutsumi, K.; Obata, Y.; Takayama, K.; Isowa, K.; Nagai, T. Permeation of several drugs through keratinized epithelial-free membrane of hamster cheek pouch. Int. J. Pharm. 1999, 177, 7–14. [Google Scholar] [CrossRef]
  110. Koschier, F.; Kostrubsky, V.; Toole, C.; Gallo, M.A. In vitro effects of ethanol and mouthrinse on permeability in an oral buccal mucosal tissue construct. Food Chem. Toxicol. 2011, 49, 2524–2529. [Google Scholar] [CrossRef]
  111. Galey, W.R.; Lonsdale, H.K.; Nacht, S. The in vitro permeability of skin and buccal mucosa to selected drugs and tritiated water. J. Investig. Dermatol. 1976, 67, 713–717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Streisand, J.B.; Zhang, J.; Niu, S.; McJames, S.; Natte, R.; Pace, N.L. Buccal absorption of fentanyl is pH-dependent in dogs. Anesthesiology 1995, 82, 759–764. [Google Scholar] [CrossRef] [PubMed]
  113. Xue, X.Y.; Zhou, Y.; Chen, Y.Y.; Meng, J.R.; Jia, M.; Hou, Z.; Bai, H.; Mao, X.G.; Luo, X.X. Promoting effects of chemical permeation enhancers on insulin permeation across TR146 cell model of buccal epithelium in vitro. Drug Chem. Toxicol. 2012, 35, 199–207. [Google Scholar] [CrossRef] [PubMed]
  114. Jacobsen, J.; Nielsen, E.B.; Brondum-Nielsen, K.; Christensen, M.E.; Olin, H.B.; Tommerup, N.; Rassing, M.R. Filter-grown TR146 cells as an in vitro model of human buccal epithelial permeability. Eur. J. Oral Sci. 1999, 107, 138–146. [Google Scholar] [CrossRef] [PubMed]
  115. Giannola, L.I.; De Caro, V.; Giandalia, G.; Siragusa, M.G.; Campisi, G.; Florena, A.M.; Ciach, T. Diffusion of naltrexone across reconstituted human oral epithelium and histomorphological features. Eur. J. Pharm. Biopharm. 2007, 65, 238–246. [Google Scholar] [CrossRef]
  116. Adrian, C.L.; Olin, H.B.; Dalhoff, K.; Jacobsen, J. In vivo human buccal permeability of nicotine. Int. J. Pharm. 2006, 311, 196–202. [Google Scholar] [CrossRef] [PubMed]
  117. Mehta, M.; Kemppainen, B.W.; Stafford, R.G. In vitro penetration of tritium-labelled water (THO) and [3H]PbTx-3 (a red tide toxin) through monkey buccal mucosa and skin. Toxicol. Lett. 1991, 55, 185–194. [Google Scholar] [CrossRef]
  118. Squier, C.A. The permeability of oral mucosa. Crit. Rev. Oral Biol. Med. 1991, 2, 13–32. [Google Scholar] [CrossRef] [PubMed]
  119. Chilcott, R.P.; Barai, N.; Beezer, A.E.; Brain, S.I.; Brown, M.B.; Bunge, A.L.; Burgess, S.E.; Cross, S.; Dalton, C.H.; Dias, M.; et al. Inter- and intralaboratory variation of in vitro diffusion cell measurements: An international multicenter study using quasi-standardized methods and materials. J. Pharm. Sci. 2005, 94, 632–638. [Google Scholar] [CrossRef] [PubMed]
  120. Ho, N.F.H.; Raub, T.J.; Burton, P.S.; Barsuhn, C.L.; Adson, A.; Audus, K.L.; Borchardt, R.T. Quantitative approaches to delineate passive transport mechanisms in cell culture monolayers. In Transport Processes in Pharmaceutical Systems; Amidon, G., Lee, P., Topp, E., Eds.; CRC Press: Boca Raton, FL, USA, 1999; pp. 219–316. [Google Scholar]
  121. Kim, Y.H.; Ghanem, A.H.; Higuchi, W.I. Model studies of epidermal permeability. Semin. Dermatol. 1992, 11, 145–156. [Google Scholar] [PubMed]
  122. Xiang, T.; Xu, Y.; Anderson, B.D. The barrier domain for solute permeation varies with lipid bilayer phase structure. J. Membr. Biol. 1998, 165, 77–90. [Google Scholar] [CrossRef]
  123. Li, S.K.; Hao, J. Transscleral passive and iontophoretic transport: Theory and analysis. Expert Opin. Drug Deliv. 2018, 15, 283–299. [Google Scholar] [CrossRef]
  124. Adson, A.; Raub, T.J.; Burton, P.S.; Barsuhn, C.L.; Hilgers, A.R.; Audus, K.L.; Ho, N.F. Quantitative approaches to delineate paracellular diffusion in cultured epithelial cell monolayers. J. Pharm. Sci. 1994, 83, 1529–1536. [Google Scholar] [CrossRef] [PubMed]
  125. Deen, W.M. Hindered transport of large molecules in liquid-filled pores. AIChE 1987, 33, 1409–1425. [Google Scholar] [CrossRef]
  126. Hao, J.; Li, S.K. Transungual iontophoretic transport of polar neutral and positively charged model permeants: Effects of electrophoresis and electroosmosis. J. Pharm. Sci. 2008, 97, 893–905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Ren, W.; Baig, A.; Li, S.K. Passive and iontophoretic transport of fluorides across enamel in vitro. J. Pharm. Sci. 2014, 103, 1692–1700. [Google Scholar] [CrossRef] [PubMed]
  128. Wen, H.; Hao, J.; Li, S.K. Characterization of human sclera barrier properties for transscleral delivery of bevacizumab and ranibizumab. J. Pharm. Sci. 2013, 102, 892–903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Chantasart, D.; Chootanasoontorn, S.; Suksiriworapong, J.; Li, S.K. Investigation of pH influence on skin permeation behavior of weak acids using nonsteroidal anti-inflammatory drugs. J. Pharm. Sci. 2015, 104, 3459–3470. [Google Scholar] [CrossRef]
  130. Fleisher, D. Biological Transport Phenomena in the Gastrointestinal Tract: Cellular Mechanisms. In Transport Processes in Pharmaceutical Systems, 1st ed.; Amidon, G.L., Lee, P.I., Topp, E.M., Eds.; CRC Press: Boca Raton, FL, USA, 1999. [Google Scholar]
  131. Winne, D. Shift of pH-absorption curves. J. Pharmacokinet. Biopharm. 1977, 5, 53–94. [Google Scholar] [CrossRef] [PubMed]
  132. Prausnitz, M.R.; Noonan, J.S. Permeability of cornea, sclera, and conjunctiva: A literature analysis for drug delivery to the eye. J. Pharm. Sci. 1998, 87, 1479–1488. [Google Scholar] [CrossRef]
  133. Li, S.K.; Peck, K.D. Passive and iontophoretic transport through the skin polar pathway. Skin Pharmacol. Physiol. 2013, 26, 243–253. [Google Scholar] [CrossRef]
  134. Hamalainen, K.M.; Kananen, K.; Auriola, S.; Kontturi, K.; Urtti, A. Characterization of paracellular and aqueous penetration routes in cornea, conjunctiva, and sclera. Investig. Ophthalmol. Vis. Sci. 1997, 38, 627–634. [Google Scholar]
Figure 1. Assessment of permeation data variability. CV of the permeability data among different research groups were plotted against the average of the permeability data. In this case, CV was calculated using the reported mean values of the same permeant from different studies conducted by different research groups and plotted as a data point using the average of these values. CV of the data among different studies from the same research groups were also determined for comparison. In this case, CV was calculated using the reported mean values of the same permeant from different studies conducted by the same research group. Permeants in studies from different research groups are: antipyrine, atenolol, bupivacaine, buspirone, caffeine, didanosine, dideoxycytidine, estradiol, mannitol, metoprolol (2 data sets of different conditions), naltrexone, ondansetron, oxprenolol, propranolol (2 data sets of different conditions), and triamcinolone acetonide. Symbols: permeant data from two (closed diamonds), three (closed circles), and four or more (closed triangles) research groups. Permeants in studies from the same research groups are: acyclovir, antipyrine, caffeine, decitabine, diazepam, didanosine, estradiol, mannitol, morphine, nicotine (2 data sets of different conditions), triamcinolone acetonide. Symbols: permeant data from two (open diamonds), three (open circles), and four or more (open triangles) studies from the same research groups.
Figure 1. Assessment of permeation data variability. CV of the permeability data among different research groups were plotted against the average of the permeability data. In this case, CV was calculated using the reported mean values of the same permeant from different studies conducted by different research groups and plotted as a data point using the average of these values. CV of the data among different studies from the same research groups were also determined for comparison. In this case, CV was calculated using the reported mean values of the same permeant from different studies conducted by the same research group. Permeants in studies from different research groups are: antipyrine, atenolol, bupivacaine, buspirone, caffeine, didanosine, dideoxycytidine, estradiol, mannitol, metoprolol (2 data sets of different conditions), naltrexone, ondansetron, oxprenolol, propranolol (2 data sets of different conditions), and triamcinolone acetonide. Symbols: permeant data from two (closed diamonds), three (closed circles), and four or more (closed triangles) research groups. Permeants in studies from the same research groups are: acyclovir, antipyrine, caffeine, decitabine, diazepam, didanosine, estradiol, mannitol, morphine, nicotine (2 data sets of different conditions), triamcinolone acetonide. Symbols: permeant data from two (open diamonds), three (open circles), and four or more (open triangles) studies from the same research groups.
Pharmaceutics 13 01814 g001
Figure 2. Relationship between log P and log Kow for all applicable permeants in Table A1.
Figure 2. Relationship between log P and log Kow for all applicable permeants in Table A1.
Pharmaceutics 13 01814 g002
Figure 3. Relationship between log P and log Dow for all applicable permeants in Table A1.
Figure 3. Relationship between log P and log Dow for all applicable permeants in Table A1.
Pharmaceutics 13 01814 g003
Figure 4. (a) Relationship between log P and MW for all applicable permeants in Table A1. (b) Enlarged figure to evaluate the relationship between log P and MW of permeants in the 130–460 Dalton range.
Figure 4. (a) Relationship between log P and MW for all applicable permeants in Table A1. (b) Enlarged figure to evaluate the relationship between log P and MW of permeants in the 130–460 Dalton range.
Pharmaceutics 13 01814 g004
Figure 5. Relationship between log P and log Kow for permeants that are not affected by pH ionization (pH independent permeants) in Table A1.
Figure 5. Relationship between log P and log Kow for permeants that are not affected by pH ionization (pH independent permeants) in Table A1.
Pharmaceutics 13 01814 g005
Figure 6. (a) Relationship between log P and log MW of polar permeants (pH independent and log Kow < −1) and macromolecules from Table A1. (b) Comparison of the log (P/Daq) ratio and log H ratio vs. log MW of the polar permeants and macromolecules. The reference P/Daq and H values in (b) are those of a hypothetical small permeant with permeability coefficient and MW equivalent to the average values of the permeants with MW < 300 Dalton in (a). Symbols: experimental P/Daq ratio data. Lines: H ratios calculated with pore radius of 1.5 nm (dotted), 2 nm (solid), 2.5 nm (dash-dot), 3 nm (short dashes), and 4 nm (dashes) using Equation (8).
Figure 6. (a) Relationship between log P and log MW of polar permeants (pH independent and log Kow < −1) and macromolecules from Table A1. (b) Comparison of the log (P/Daq) ratio and log H ratio vs. log MW of the polar permeants and macromolecules. The reference P/Daq and H values in (b) are those of a hypothetical small permeant with permeability coefficient and MW equivalent to the average values of the permeants with MW < 300 Dalton in (a). Symbols: experimental P/Daq ratio data. Lines: H ratios calculated with pore radius of 1.5 nm (dotted), 2 nm (solid), 2.5 nm (dash-dot), 3 nm (short dashes), and 4 nm (dashes) using Equation (8).
Pharmaceutics 13 01814 g006aPharmaceutics 13 01814 g006b
Table 1. Comparison between the permeability coefficients of porcine buccal tissue and other species and engineered tissues. Data are shown as log P (P in cm/s).
Table 1. Comparison between the permeability coefficients of porcine buccal tissue and other species and engineered tissues. Data are shown as log P (P in cm/s).
PermeantPorcine aBovineCanineHamsterMonkeyHumanEngineered TissueRef b
Acebutolol−7.40, −7.57 −5.46 d[34]
Alprenolol−5.70 −4.47 d[34]
Atenolol−7.55 to −6.72 −6.41 e, −5.68 d[34,39]
Caffeine−5.57 to −4.63 −2.21 −4.96 e, −6.11 f[39,109,110]
Cathine−5.50 −5.00 e[39]
Cathinone−5.52 −4.92 e[39]
Estradiol−4.54, −4.29 −4.88 [111]
Fentanyl−5.15 −4.54 [112]
Galantamine−4.93 −4.70 d[68]
Insulin−8.18 −5.92 d, −7.10 d[70,113]
Labetalol−7.70, −6.26 −5.11 d[34]
Lidocaine−6.57 to −4.77 −2.13 [109]
Mannitol−7.00 to −5.60 −6.51−6.62, −6.62−5.39 d, −5.66 d[34,78,114]
Metoprolol−8.60 to −4.97 −5.10−4.47 d[34,78]
Morphine−5.88, −5.72−6.60 [84]
Naltrexone−5.28, −4.76 −5.07 c,f, −4.98 d[86,115]
Nicotine−7.90 to −4.50 −3.77 c [116]
Norephedrine−5.67 −5.01 e[39]
Oxprenolol−6.05, −5.52 −4.40 d[34]
Pindolol−6.92, −6.70 −4.51 d[34]
Propranolol−5.89 to −4.85 −5.38 d[34]
Tertatolol−6.00 −4.57 d[34]
Testosterone−5.96 −4.72−5.74−4.70 d[34]
Timolol−6.52 −4.62 d[34]
Verapamil−4.60 −2.52 [109]
Water−5.09, −4.94 −4.29 −5.77, −4.62−6.02−4.28 d[34,111,117,118]
a Permeability value from Table A1. b References listed are for buccal tissues from other species and engineered tissues. References for porcine buccal tissue are listed in Table A1. c Multiple permeability values are available in the reference and the average value of log P was used. d TR146 cell culture model. e Caco-2 cell model. f EpiOral model.
Table 2. Regression analysis results of log P with log Dow and MW as independent variables (first three rows) and log (P/Dow) with log Kow and MW as independent variables (last three rows), using the data of all permeants and then permeants with log Dow > −1 and > 0. The parameters are defined in Equations (6) and (10). The values presented are the least squares means and standard errors of the parameters.
Table 2. Regression analysis results of log P with log Dow and MW as independent variables (first three rows) and log (P/Dow) with log Kow and MW as independent variables (last three rows), using the data of all permeants and then permeants with log Dow > −1 and > 0. The parameters are defined in Equations (6) and (10). The values presented are the least squares means and standard errors of the parameters.
ConditionacconstantR2
log P vs. log Dow and MW; all data; n = 1150.29 ± 0.05−0.0008 ± 0.0009 −5.5 ± 0.30.222
log P vs. log Dow and MW; log Dow > −1; n = 880.37 ± 0.08−0.0021 ± 0.0011−5.3 ± 0.30.183
log P vs. log Dow and MW; log Dow > 0; n = 620.22 ± 0.10−0.0007 ± 0.0011−5.4 ± 0.30.072
log (P/Dow) vs. log Kow and MW; all data; n = 1150.44 ± 0.05−0.0014 ± 0.0009−4.8 ± 0.30.588
log (P/Dow) vs. log Kow and MW; log Dow > −1; n = 880.60 ± 0.09−0.0031 ± 0.0013−4.7 ± 0.30.382
log (P/Dow) vs. log Kow and MW; log Dow > 0; n = 620.64 ± 0.12−0.0027 ± 0.0014−5.0 ± 0.40.279
Table 3. Regression analysis results of log P with log Kow and MW as independent variables, using the data of pH independent permeants and then with the conditions of log Kow > −1 and >0. The parameters are defined in Equation (11). The values presented are the least squares means and standard errors of the parameters.
Table 3. Regression analysis results of log P with log Kow and MW as independent variables, using the data of pH independent permeants and then with the conditions of log Kow > −1 and >0. The parameters are defined in Equation (11). The values presented are the least squares means and standard errors of the parameters.
ConditionacconstantR2
log P vs. log Kow and MW; all data; n = 440.16 ± 0.04−0.0012 ± 0.0007−5.2 ± 0.20.246
log P vs. log Kow and MW; log Kow > −1; n = 280.17 ± 0.07−0.0031 ± 0.0009−4.8 ± 0.20.325
log P vs. log Kow and MW; log Kow > 0; n = 180.20 ± 0.06−0.0033 ± 0.0006−4.8 ± 0.20.615
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wanasathop, A.; Patel, P.B.; Choi, H.A.; Li, S.K. Permeability of Buccal Mucosa. Pharmaceutics 2021, 13, 1814. https://doi.org/10.3390/pharmaceutics13111814

AMA Style

Wanasathop A, Patel PB, Choi HA, Li SK. Permeability of Buccal Mucosa. Pharmaceutics. 2021; 13(11):1814. https://doi.org/10.3390/pharmaceutics13111814

Chicago/Turabian Style

Wanasathop, Apipa, Priya B Patel, Hyojin A. Choi, and S. Kevin Li. 2021. "Permeability of Buccal Mucosa" Pharmaceutics 13, no. 11: 1814. https://doi.org/10.3390/pharmaceutics13111814

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