Next Article in Journal
Antioxidant Efficacy and “In Vivo” Safety of a Bentonite/Vitamin C Hybrid
Next Article in Special Issue
Advance and Challenges in the Treatment of Skin Diseases with the Transdermal Drug Delivery System
Previous Article in Journal
Lights and Dots toward Therapy—Carbon-Based Quantum Dots as New Agents for Photodynamic Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Immobilized Keratin HPLC Stationary Phase—A Forgotten Model of Transdermal Absorption: To What Molecular and Biological Properties Is It Relevant?

by
Anna Weronika Sobańska
* and
Elżbieta Brzezińska
Department of Analytical Chemistry, Faculty of Pharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
*
Author to whom correspondence should be addressed.
Pharmaceutics 2023, 15(4), 1172; https://doi.org/10.3390/pharmaceutics15041172
Submission received: 28 February 2023 / Revised: 28 March 2023 / Accepted: 4 April 2023 / Published: 7 April 2023
(This article belongs to the Special Issue Transdermal/Dermal Drug Delivery System)

Abstract

:
Chromatographic retention data collected on immobilized keratin (KER) or immobilized artificial membrane (IAM) stationary phases were used to predict skin permeability coefficient (log Kp) and bioconcentration factor (log BCF) of structurally unrelated compounds. Models of both properties contained, apart from chromatographic descriptors, calculated physico-chemical parameters. The log Kp model, containing keratin-based retention factor, has slightly better statistical parameters and is in a better agreement with experimental log Kp data than the model derived from IAM chromatography; both models are applicable primarily to non-ionized compounds.Based on the multiple linear regression (MLR) analyses conducted in this study, it was concluded that immobilized keratin chromatographic support is a moderately useful tool for skin permeability assessment.However, chromatography on immobilized keratin may also be of use for a different purpose—in studies of compounds’ bioconcentration in aquatic organisms.

1. Introduction

Many chemicals enter the human body through the skin. Transdermal absorption is an important route of drugs’ administration, and it is also very important in the context of environmental toxicology, since undesired xenobiotics are often absorbed transdermally. The skin permeability coefficient Kp is defined according to Equation (1):
K p = K m D h
where: Km—the partition coefficient between the stratum corneum and the vehicle; D—the effective compound’s diffusion coefficient through the stratum corneum; h—the diffusional pathlength.
The experimental values of skin permeability coefficients are measured in vivo (on human volunteers), ex vivo (on excised human skin), or on animal models [1], but such data are difficult to obtain due to ethical and financial problems, and the results of experiments in this area are often inconsistent due to variations in properties of different skin samples, even taken from the same human or animal.
Apart from skin absorption, an important property of compounds of environmental concern is their bioconcentration factor in aquatic organisms (BCF). The bioconcentration factor is the ratio of the chemical concentration in the organism (CB) and water (Cw), accounting for the absorption via the respiratory route (e.g., gills) and skin. It is used to assess the bioaccumulation potential of compounds [2], especially in the absence of their bioaccumulation factor (BAF),which accounts for dietary, dermal, and respiratory exposures. According to different regulatory agencies, different criteria of bioaccumulation apply: bioaccumulative compounds have BCF > 5000 or BCF > 2000 [3]. In the absence of BAF or BCF data, lipophilicity measured as the octanol-water partition coefficient Kow is used to assess the compounds’ ability to bioaccumulate; if this is the case, the log Kow threshold for bioaccumulative compounds is 5 [3,4], 4.5 [5], or 3.3 [6]. Measured and estimated bioaccumulation data are also used to assign chemicals to three bioaccumulation categories: not significantly bioaccumulative (BCF or BAF < 1000), bioaccumulative (BCF or BAF between 1000 and 5000), and highly bioaccumulative (BCF or BAF > 5000) [7].
The ethical and financial problems related to BCF determination are similar to those encountered during Kp measurements. In in vivo experiments, the need to use human volunteers or lab animals, as well as the experiment timing, are the main limitations, and, for this reason, both Kp and BCF are frequently assessed in vitro (using cell/tissue assays or non-cell models based on chromatography or electrochromatography) or in silico (calculations that can provide valuable information even without the access to compounds’ samples) [8,9,10].
Biomimetic chromatography essentially involves the application of stationary phases, containing proteins or phospholipids, or mobile phases, including micelles or microemulsions [11,12,13,14]. The components of biomimetic chromatographic systems (stationary or mobile phases) are designed to mimic some elements or functions of biomembranes and the interactions between these components and studied molecules resemble transport and partition phenomena encountered in a living being.
Immobilized artificial membrane (IAM) chromatography, with stationary phases containing adsorbed or covalently bound phosphatidylcholine (or, more recently, sphyngomyelin) groups, is used in modern lipohilicity studies, as well as in investigations of compounds’ affinity for phospholipids, related to many biological properties of solutes [15,16,17]. Chromatography on immobilized protein stationary phases was originally developed to separate enantiomers [18]; apart from that, some protein-based stationary phases simulate the interaction between a molecule and main plasma proteins, such as human serum albumin (HSA) [19,20,21,22] or α1 acid glycoprotein (AGP) [21,23,24,25]. Retention data obtained from chromatography in biomimetic systems are used to predict ADME (absorption, distribution, metabolism, and excretion) properties of compounds in early drug discovery phases [11,26], as well as their environmental impact—mobility in soil, bioconcentration/bioaccumulation, or aquatic toxicity [27,28,29,30,31]. Elements of natural biomembranes, incorporated in chromatographic systems used in pharmacokinetic studies, include also cholesterol or amide moieties [32,33].
Chromatographic descriptors have been used in skin permeability studies for many years, and separation (chromatographic or electrochromatographic) techniques used in these studies are liquid chromatography (HPLC or TLC), biopartitioning micellar chromatography, micellar electrokinetic chromatography, liposome electrokinetic chromatography, and two-dimensional gas chromatography (GC × GC) [34,35,36,37,38,39,40,41,42,43,44,45,46,47].
The relationships between the IAM chromatographic retention factor (kIAM) and the skin permeability coefficient have been studied most frequently for small groups of compounds (n = 10 to 32), and the resulting dependencies are mostly univariate (linear or quadratic) [35,36,39,41], the exceptions being the studies in which additional variables, e.g., McGowan’s characteristic volume V or the octanol-water partition coefficient log Kow [33,35,36] were incorporated. In our earlier study [48] conducted for a large group of structurally unrelated compounds (n = 160), we demonstrated that log kIAM accounts for ca. 46% of total log Kp variability, and the parameters whose contribution to log Kp predictions is also significant are polar surface area (PSA) or polarizability (α).
Bioconcentration of compounds in aquatic organisms can be studied in vitro using descriptors derived from HPLC chromatography on C18, C8, C2, and phenyl-bonded silica sorbents (aromatic hydrocarbons [49]), C18 and cyanopropyl- and phenyl-bonded silica (aromatic hydrocarbons, alkylbenzenes, chlorinated benzenes, phthalates, nitroaromatics, phenols, and aniline [50]), and RP-18 TLC (organic sunscreens and cosmetic preservatives [51]).
More recently, the bioconcentration of compounds in aquatic organisms has been investigated using chromatography on IAM stationary phases, developed initially to mimic molecule–biomembrane interactions in ADME studies [31,52]. Earlier research pointed to the importance of additional parameters, incorporated alongside log kIAM: (i) a biodegradation estimate, BioWin5, calculated using the EPISuiteTM software and (to a lesser extent) topological polar surface area (TPSA) [52]; (ii) TPSA—the fraction of sp3 carbon atoms (FCsp3) and hydrogen bond donor count (#HD) [31].
Turowski and Kaliszan postulated that predicting skin permeability of compounds should be based on molecules’ lipophilicity and interactions with keratin, which is an important constituent of the outmost layer of the epidermis [34]. An immobilized keratin-based stationary phase, developed by Turowski and Kaliszan, was initially proposed to be an in vitro tool in investigations of solutes′ skin permeability (log Kp) [34]. However, it was discovered that the retention factor obtained on this sorbent (log kKER) is not a sufficiently good predictor of skin permeability coefficient, and it cannot be used as a sole descriptor in log Kp models. Turowski and Kaliszan reported that this descriptor can be combined with the chromatographic retention factor obtained by immobilized artificial membrane chromatography (log kIAM), and the results of log Kp predictions using multiple linear regression (MLR) models satisfy (Equation (2)):
log Kp = −6.56 + 1.92 log kIAM − 1.04 log kKER (n = 17, R2 = 0.86)
Turowski and Kaliszan concluded that skin permeability increases with the lipophilicity of solutes (encoded primarily by log kIAM) and decreases with their affinity for keratin (expressed as log kKER). Unfortunately, the model they proposed (Equation (2)) requires two sets of chromatographic data, obtained on different stationary phases, this being the likely reason why the immobilized keratin stationary phase they proposed has never become widely popular and, to the best of our knowledge, it is not commercially available.
In this study, a novel application of immobilized keratin stationary phases developed by Turowski and Kaliszan is proposed, and chromatography on immobilized keratin sorbent is used to model compounds’ bioconcentration in aquatic organisms.

2. Materials and Methods

2.1. IAM and Immobilized Keratin Chromatography

The chromatographic retention factors for the compounds analyzed in this study (Table 1) were taken from [34]. They were obtained on: (i) an IAM.PC.MG HPLC column purchased from Regis (150 × 4.6 mm, particle diameter 12 μm, pore diameter 300 Å) with a phosphate buffer (pH 6.0), including acetonitrile (95:5 v/v) mobile phase (flow rate—1 mL min−1); (ii) physically immobilized keratin sorbent with pH 4.2 phosphate buffer as a mobile phase (column dimensions—125 × 4 mm; flow rate—1 mL min−1). The mobile phase used in keratin chromatography (pH 4.2 buffer) was selected on the basis of QSRR studies as giving the “best” relationship between log kKER and structural descriptors (molecular weight and dipole moment) [34].

2.2. Calculated Molecular Descriptors

Molecular weight (Mw), heavy atom count (#HvAt), aromatic heavy atom count (#ArHvAt), fraction of sp3 carbons (FCsp3), rotatable bond count (#FRB), hydrogen donor count (#HD), hydrogen acceptor count (#HA), molecular refractivity (MR), aqueous solubility (log S), and topological polar surface area (TPSA) were calculated using Swiss ADME software available freely on-line [53]. The octanol–water partition coefficient (log Kow) was predicted using EpiSuite [54]. Total counts of nitrogen and oxygen atoms (N + O) were calculated manually on the basis of compounds’ molecular formulas (Table 1).

2.3. Reference Values of Skin Permeability Coefficient (log Kp) and Bioconcentration Factor (log BCF)

The experimentally determined values of log Kp and log BCF are available for only some compounds within the studied group. For this reason, the models of skin permeability and bioconcentration factor, involving chromatographic and calculated descriptors, were generated and validated using log Kp and log BCF values obtained in silico with the EpiSuite v. 4.1 software (log KpEPI—DERMWIN v. 2.02 and log BCFEPI—BCFBAF v. 3.02 modules, respectively), recommended by the US Environmental Protection Agency [54,55] and tested on sub-groups of solutes whose experimental log Kp or log BCF values are known (log Kpexp, log BCFexp) [56,57]. The estimation methodology used by DERMWIN is based on an algorithm developed by Potts [58], and the estimations provided by BCFBAF are based on methodology developed by Meylan [59] and Arnot and Gobas [3]. The values of log KpEPI and log BCFEPI obtained using EpiSuite are given in Table 2 and Table 3.

2.4. Statistical Tools

Multiple linear regression (MLR) models were generated using Statistica v. 13 by StatSoft Polska, Kraków, Poland, and this refers to the stepwise forward regression mode.
The models considered in this study were evaluated using the following procedures:
  • Cross-validation was performed, with n compounds from the initial training set split into 2 subsets, one of which was used to train a new model and the remaining one to test it. After cross-validation, the RMSEP (root mean squared error of prediction) for the particular N-compound test subset was calculated as follows (Equation (3)):
R M S E P = i = 1 N y i p r e d y i r e f 2 N
  • Comparison of the predicted log Kppred and log BCFpred values (calculated for the compounds, whose experimental log Kpexp and log BCFexp data are available) was performed, and these data were analyzed using the squared coefficient of determination (R2exp).

3. Results

3.1. Keratin vs. IAM HPLC Skin Permeability Models

In this study, we compared the log Kp models obtained using log kIAM and TPSA (Equation (4)) with the models including log kKER as a chromatographic parameter (Equation (5)).
log Kp = −5.61 (±0.24) + 0.68 (±0.17) log kIAM − 0.014 (±0.005) TPSA
(n = 32, R2 = 0.63, R2adj. = 0.63, R2exp = 0.72, F = 25.1, p< 0.01)
log Kp = −2.56 (±0.83) +1.74 (±0.38) log kKER − 0.011 (±0.008) Mw
− 0.22 (±0.11) #ArHvAt − 0.014 (±0.005) TPSA
(n = 32, R2 = 0.68, R2adj. = 0.63, R2exp= 0.73, F = 14.3, p< 0.01)
It was observed that neither Equation (4), nor (5), gives satisfying results of log Kp predictions for relatively strongly ionized solutes (compounds 14, 16, 23, and 27); when these compounds were excluded from the analysis, Equations (6) and (7) were obtained for a group of 28 neutral, basic, or weakly acidic compounds (Figure 1 and Figure 2, Table 2).
log Kp = −5.70 + 0.81 (±0.17) log kIAM− 0.015 (±0.004) TPSA
(n = 28, R2 = 0.80, R2adj. = 0.78, R2exp = 0.73, F = 49.7, p< 0.01)
log Kp = −2.73 (±0.54) +1.80 (±0.26) log kKER − 0.015 (±0.003) Mw + 0.13 (±0.05) #HvAt − 0.27 (±0.07) #ArHvAt − 0.020 (±0.004) TPSA
(n = 28, R2 = 0.85, R2adj. = 0.81, R2exp = 0.79, F = 24.8, p< 0.01)
The likely reason for such discrepancies between the predicted (Equations (4) and (5)) and reference values of log Kp for relatively strongly ionizable compounds is that the reference model has also its limitations: it overestimates the results for very hydrophilic molecules, underestimates the values for non-hydrogen bonding solutes, and fails for extremely lipophilic compounds or solutes having a very high tendency to hydrogen bonding [60,61,62].
At this point, the group of 28 studied compounds was divided into two subsets: a training set (1 to 20) and a test set (21 to 28). Equations (8) and (9) generated for the training set, and containing the same sets of independent variables as Equations (6) and (7), are as follows (Table 2):
log Kp = −6.09 (±0.27) + 0.94 (±0.17) log kIAM − 0.0073 (±0.005) TPSA
(n = 20, R2 = 0.80, R2adj. = 0.78, RMSEP = 0.51, F = 34.2, p< 0.01)
log Kp = −1.93 (±0.54) +1.85 (±0.28) log kKER − 0.017 (±0.003) Mw + 0.15 (±0.05) #HvAt − 0.37 (±0.11) #ArHvAt − 0.021 (±0.004) TPSA
(n = 20, R2 = 0.87, R2adj. = 0.83, RMSEP = 0.44, F = 19.0, p< 0.01)

3.2. Keratin HPLC Models of Bioconcentration Factor

According to our earlier research, the bioconcentration factor log BCF can be predicted using log kIAM and two additional parameters: FCsp3 and TPSA [31]. The predictive potential of Equation (10) (Figure 3) is compared to that of a model based on chromatographic retention factors obtained using immobilized keratine as a stationary phase (Equation (11), Figure 4).
log BCF = 0.79 (±0.11) + 0.62 (±0.07) log kIAM + 1.53 (±0.31) FCsp3
− 0.0046 (±0.0021) TPSA
(n = 32, R2 = 0.87, R2adj. = 0.86, R2exp = 0.41, F = 63.9, p < 0.01)
log BCF = 1.23 (±0.34) + 0.70 (±0.15) log kKER − 0.18 (±0.05)
#ArHvAt + 0.039 (±0.006) MR − 0.017 (±0.002) TPSA
(n = 32, R2 = 0.88, R2adj. = 0.86, R2exp = 0.69, F = 50.3, p < 0.01)
At this point, the group of 32 studied compounds was divided into two subsets: a training set (1 to 20) and a test set (21 to 32). Equations (12) and (13) generated for the training set, and containing the same sets of independent variables as Equations (10) and (11) are as follows:
log BCF = 0.46 (±0.15) + 0.75 (±0.09) log kIAM + 1.84 (±0.34) FCsp3
+ 0.0010 (±0.0028) TPSA
(n = 20, R2 = 0.93, R2adj. = 0.92, RMSEP = 0.36, F = 72.2, p < 0.01)
log BCF = 1.59 (±0.57) + 0.85 (±0.20) log kKER − 0.22 (±0.08)
#ArHvAt + 0.037 (±0.007) MR − 0.018 (±0.003) TPSA
(n = 20, R2 = 0.90, R2adj. = 0.88, RMSEP = 0.23, F = 35.1, p < 0.01)

4. Discussion

In our study, we investigated the possibility of using log kKER in skin permeability models, alongside additional descriptors that were either not considered or not available when the keratin stationary phase was originally developed. We studied correlations between log kKER and the key physico-chemical properties associated with compounds’ ability to cross biological barriers (Table 4) and discovered that log kKER encodes primarily lipophilicity (log Kow) and aqueous solubility (log S), which are important factors governing the ability of compounds to cross the skin barrier, but the correlations are moderate.
Predictive models of log Kp, involving retention parameters obtained on immobilized keratin (Equations (7) and (9)), have similar (or, in fact, slightly better) statistical parameters compared to those reported for models based on IAM chromatography (Equations (6) and (8)). Log Kp values predicted using Equation (7) are in a slightly closer agreement with experimental log Kpexp data available for a subset of 18 compounds than those calculated using Equation (6). It must be noted, however, that, in the process of descriptors’ selection by forward stepwise regression, chromatographic parameters log kKER and log kIAM behave differently. Log kIAM (Equation (6)) is selected first, and it accounts for ca. 66% of total log Kp variability; log kKER (Equation (7)) is selected second (after TPSA), and it accounts for just 16% of total log Kp variability.
The significance of log kKER as an independent variable is much higher in models of bioconcentration factor log BCF. In Equation (11), log kKER is the most important independent variable, accounting for 39% of total log Kp variability; further variables (selected as follows: TPSA, MR, and #ArHvAt) account for 24, 18, and 7% of total log Kp variability, respectively. In the IAM chromatography-based model of log BCF (Equation (10)), log kIAM accounts for 73%, and other independent variables (FCsp3 and TPSA) account for 12 and 2% of total log Kp variability, respectively. The keratin chromatographic retention-based model (11) has statistical parameters similar to those of Equation (10), derived from IAM chromatography; however, Equation (11) seems to fit the experimental data (log BCFexp) reported for a subset of 10 compounds better than Equation (10).

5. Conclusions

Immobilized keratine-based chromatographic stationary phase was developed in the late 1990s to help in in vitro investigations of compounds’ transdermal absorption. A new model of a skin permeability coefficient was developed in the current study, which involves the chromatographic retention factor measured on the immobilized keratine sorbent (log kKER) and four additional independent variables (Equation (7)). This model has slightly better statistical parameters and is in a better agreement with experimental log Kp data than the model derived from IAM chromatography (Equation (6)); both models are applicable primarily to non-ionized compounds (with carboxylic acids removed from Equations (4) and (5)). Based on the MLR analyses conducted in this study, it was concluded that immobilized keratin chromatographic support is a moderately useful tool for skin permeability assessment. However, similarly to IAM chromatography in the past, chromatography on immobilized keratin may serve a different purpose; designed for applications in pharmacokinetic studies, it may also be of use in the realm of environmental science, in studies of compounds’ bioconcentrations in aquatic organisms.

Author Contributions

Conceptualization, A.W.S.; methodology, A.W.S. and E.B.; investigation, A.W.S.; writing—original draft preparation, A.W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research study was supported by an internal grant of the Medical University of Lodz, no. 503/3-016-03/503-31-001.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data generated in this study can be found in this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Todo, H. Transdermal Permeation of Drugs in Various Animal Species. Pharmaceutics 2017, 9, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Weisbrod, A.V.; Burkhard, L.P.; Arnot, J.; Mekenyan, O.; Howard, P.H.; Russom, C.; Boethling, R.; Sakuratani, Y.; Traas, T.; Bridges, T.; et al. Workgroup Report: Review of Fish Bioaccumulation Databases Used to Identify Persistent, Bioaccumulative, Toxic Substances. Environ. Health Perspect 2007, 115, 255–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Arnot, J.A.; Gobas, F.A. A Review of Bioconcentration Factor (BCF) and Bioaccumulation Factor (BAF) Assessments for Organic Chemicals in Aquatic Organisms. Environ. Rev. 2006, 14, 257–297. [Google Scholar] [CrossRef]
  4. Arnot, J.A.; Gobas, F.A.P.C. A Generic QSAR for Assessing the Bioaccumulation Potential of Organic Chemicals in Aquatic Food Webs. QSAR Comb. Sci. 2003, 22, 337–345. [Google Scholar] [CrossRef]
  5. Grisoni, F.; Consonni, V.; Villa, S.; Vighi, M.; Todeschini, R. QSAR Models for Bioconcentration: Is the Increase in the Complexity Justified by More Accurate Predictions? Chemosphere 2015, 127, 171–179. [Google Scholar] [CrossRef]
  6. Chmiel, T.; Mieszkowska, A.; Kempińska-Kupczyk, D.; Kot-Wasik, A.; Namieśnik, J.; Mazerska, Z. The Impact of Lipophilicity on Environmental Processes, Drug Delivery and Bioavailability of Food Components. Microchem. J. 2019, 146, 393–406. [Google Scholar] [CrossRef]
  7. Costanza, J.; Lynch, D.G.; Boethling, R.S.; Arnot, J.A. Use of the Bioaccumulation Factor to Screen Chemicals for Bioaccumulation Potential. Environ. Toxicol. Chem. 2012, 31, 2261–2268. [Google Scholar] [CrossRef]
  8. Wanat, K. Biological Barriers, and the Influence of Protein Binding on the Passage of Drugs across Them. Mol. Biol. Rep. 2020, 47, 3221–3231. [Google Scholar] [CrossRef] [Green Version]
  9. Alonso, C.; Carrer, V.; Espinosa, S.; Zanuy, M.; Córdoba, M.; Vidal, B.; Domínguez, M.; Godessart, N.; Coderch, L.; Pont, M. Prediction of the Skin Permeability of Topical Drugs Using In Silico and In Vitro Models. Eur. J. Pharm. Sci. 2019, 136, 104945. [Google Scholar] [CrossRef]
  10. Bertato, L.; Chirico, N.; Papa, E. Predicting the Bioconcentration Factor in Fish from Molecular Structures. Toxics 2022, 10, 581. [Google Scholar] [CrossRef]
  11. Valko, K. Application of Biomimetic HPLC to Estimate In Vivo Behavior of Early Drug Discovery Compounds. Future Drug Discov. 2019, 1, FDD11. [Google Scholar] [CrossRef] [Green Version]
  12. Valko, K.L. Biomimetic Chromatography—A Novel Application of the Chromatographic Principles. Anal. Sci. Adv. 2022, 3, 146–153. [Google Scholar] [CrossRef]
  13. Russo, G.; Grumetto, L.; Szucs, R.; Barbato, F.; Lynen, F. Determination of In Vitro and In Silico Indexes for the Modeling of Blood-Brain Barrier Partitioning of Drugs via Micellar and Immobilized Artificial Membrane Liquid Chromatography. J. Med. Chem. 2017, 60, 3739–3754. [Google Scholar] [CrossRef] [Green Version]
  14. Ulenberg, S.; Ciura, K.; Georgiev, P.; Pastewska, M.; Ślifirski, G.; Król, M.; Herold, F.; Bączek, T. Use of Biomimetic Chromatography and In Vitro Assay to Develop Predictive GA-MLR Model for Use in Drug-Property Prediction among Anti-Depressant Drug Candidates. Microchem. J. 2022, 175, 107183. [Google Scholar] [CrossRef]
  15. Ermondi, G.; Vallaro, M.; Caron, G. Learning How to Use IAM Chromatography for Predicting Permeability. Eur. J. Pharm. Sci. 2018, 114, 385–390. [Google Scholar] [CrossRef]
  16. Verzele, D.; Lynen, F.; Vrieze, M.D.; Wright, A.G.; Hanna-Brown, M.; Sandra, P. Development of the First Sphingomyelin Biomimetic Stationary Phase for Immobilized Artificial Membrane (IAM) Chromatography. Chem. Commun. 2012, 48, 1162–1164. [Google Scholar] [CrossRef]
  17. Ciura, K.; Kovačević, S.; Pastewska, M.; Kapica, H.; Kornela, M.; Sawicki, W. Prediction of the Chromatographic Hydrophobicity Index with Immobilized Artificial Membrane Chromatography Using Simple Molecular Descriptors and Artificial Neural Networks. J. Chromatogr. A 2021, 1660, 462666. [Google Scholar] [CrossRef]
  18. Haginaka, J.; Kanasugi, N. Enantioselectivity of Bovine Serum Albumin-Bonded Columns Produced with Isolated Protein Fragments. II. Characterization of Protein Fragments and Chiral Binding Sites. J. Chromatogr. A 1997, 769, 215–223. [Google Scholar] [CrossRef]
  19. Kim, H.S.; Wainer, I.W. Rapid Analysis of the Interactions between Drugs and Human Serum Albumin (HSA) Using High-Performance Affinity Chromatography (HPAC). J. Chromatogr. B 2008, 870, 22–26. [Google Scholar] [CrossRef] [Green Version]
  20. Ishii, T.; Minoda, K.; Bae, M.-J.; Mori, T.; Uekusa, Y.; Ichikawa, T.; Aihara, Y.; Furuta, T.; Wakimoto, T.; Kan, T.; et al. Binding Affinity of Tea Catechins for HSA: Characterization by High-Performance Affinity Chromatography with Immobilized Albumin Column. Mol. Nutr. Food Res. 2009, 54, 816–822. [Google Scholar] [CrossRef]
  21. Valko, K.; Nunhuck, S.; Bevan, C.; Abraham, M.H.; Reynolds, D.P. Fast Gradient HPLC Method to Determine Compounds Binding to Human Serum Albumin. Relationships with Octanol/Water and Immobilized Artificial Membrane Lipophilicity. J. Pharm. Sci. 2003, 92, 2236–2248. [Google Scholar] [CrossRef] [PubMed]
  22. Chrysanthakopoulos, M.; Giaginis, C.; Tsantili-Kakoulidou, A. Retention of Structurally Diverse Drugs in Human Serum Albumin Chromatography and Its Potential to Simulate Plasma Protein Binding. J. Chromatogr. A 2010, 1217, 5761–5768. [Google Scholar] [CrossRef] [PubMed]
  23. Anguizola, J.; Bi, C.; Koke, M.; Jackson, A.; Hage, D.S. On-Column Entrapment of Alpha1-Acid Glycoprotein for Studies of Drug-Protein Binding by High-Performance Affinity Chromatography. Anal. Bioanal. Chem. 2016, 408, 5745–5756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Xuan, H.; Hage, D.S. Immobilization of A1-Acid Glycoprotein for Chromatographic Studies of Drug–Protein Binding. Anal. Biochem. 2005, 346, 300–310. [Google Scholar] [CrossRef]
  25. Filipic, S.; Ruzic, D.; Vucicevic, J.; Nikolic, K.; Agbaba, D. Quantitative Structure-Retention Relationship of Selected Imidazoline Derivatives on A1-Acid Glycoprotein Column. J. Pharm. Biomed. Anal. 2016, 127, 101–111. [Google Scholar] [CrossRef]
  26. Vallianatou, T.; Tsopelas, F.; Tsantili-Kakoulidou, A. Prediction Models for Brain Distribution of Drugs Based on Biomimetic Chromatographic Data. Molecules 2022, 27, 3668. [Google Scholar] [CrossRef]
  27. Stergiopoulos, C.; Tsopelas, F.; Valko, K.; Ochsenkühn-Petropoulou, M. The Use of Biomimetic Chromatography to Predict Acute Aquatic Toxicity of Pharmaceutical Compounds. Toxicol. Environ. Chem. 2021, 104, 1–9. [Google Scholar] [CrossRef]
  28. Andrić, F.; Šegan, S.; Tešić, Ž.; Milojković-Opsenica, D. Chromatographic Methods in Determination of the Soil–Water Partition Coefficient. J. Liq. Chromatogr. Relat. Technol. 2016, 39, 249–256. [Google Scholar] [CrossRef]
  29. Hidalgo-Rodríguez, M.; Fuguet, E.; Ràfols, C.; Rosés, M. Performance of Chromatographic Systems to Model Soil–Water Sorption. J. Chromatogr. A 2012, 1252, 136–145. [Google Scholar] [CrossRef]
  30. Sobańska, A.W. Immobilized Artificial Membrane-Chromatographic and Computational Descriptors in Studies of Soil-Water Partition of Environmentally Relevant Compounds. Env. Sci. Pollut. Res. 2022, 30, 6192–6200. [Google Scholar] [CrossRef]
  31. Sobańska, A.W. Affinity of Compounds for Phosphatydylcholine-Based Immobilized Artificial Membrane—A Measure of Their Bioconcentration in Aquatic Organisms. Membranes 2022, 12, 1130. [Google Scholar] [CrossRef]
  32. Farsa, O. Chromatographic Behaviour Predicts the Ability of Potential Nootropics to Permeate the Blood-Brain Barrier. Sci. Pharm. 2013, 81, 81–91. [Google Scholar] [CrossRef] [Green Version]
  33. Grooten, Y.; Mangelings, D.; Van der Heyden, Y. Predicting Skin Permeability of Pharmaceutical and Cosmetic Compounds Using Retention on Octadecyl, Cholesterol-Bonded and Immobilized Artificial Membrane Columns. J. Chromatogr. A 2022, 1676, 463271. [Google Scholar] [CrossRef]
  34. Turowski, M.; Kaliszan, R. Keratin Immobilized on Silica as a New Stationary Phase for Chromatographic Modelling of Skin Permeation. J. Pharm. Biomed. Anal. 1997, 15, 1325–1333. [Google Scholar] [CrossRef]
  35. Barbato, F.; Cappello, B.; Miro, A.; La Rotonda, M.; Quaglia, F. Chromatographic Indexes on Immobilized Artificial Membranes for the Prediction of Transdermal Transport of Drugs. Il Farm. 1998, 53, 661–665. [Google Scholar] [CrossRef]
  36. Hidalgo-Rodríguez, M.; Soriano-Meseguer, S.; Fuguet, E.; Ràfols, C.; Rosés, M. Evaluation of the Suitability of Chromatographic Systems to Predict Human Skin Permeation of Neutral Compounds. Eur. J. Pharm. Sci. 2013, 50, 557–568. [Google Scholar] [CrossRef]
  37. Jevric, L.R.; Podunavac Kuzmanovic, S.O.; Svarc Gajic, J.V.; Kovacevic, S.; Jovanovic, B.Z. RP-HPTLC Retention Data in Correlation with the In-Silico ADME Properties of a Series of s-Triazine Derivatives. Iran. J. Pharm. Res. 2014, 13, 1203–1211. [Google Scholar] [CrossRef]
  38. Kovačević, S.; Jevrić, L.R.; Podunavac Kuzmanović, S.O.; Lončar, E.S. Prediction of In Silico ADME Properties of 1,2-O-Isopropylidene Aldohexose Derivatives. Iran. J. Pharm. Res. 2014, 13, 899–907. [Google Scholar] [CrossRef]
  39. Lazaro, E.; Rafols, C.; Abraham, M.H.; Rosés, M. Chromatographic Estimation of Drug Disposition Properties by Means of Immobilized Artificial Membranes (IAM) and C18 Columns. J. Med. Chem. 2006, 49, 4861–4870. [Google Scholar] [CrossRef]
  40. Martínez-Pla, J.J.; Martín-Biosca, Y.; Sagrado, S.; Villanueva-Camañas, R.M.; Medina-Hernández, M.J. Evaluation of the pH Effect of Formulations on the Skin Permeability of Drugs by Biopartitioning Micellar Chromatography. J. Chromatogr. A 2004, 1047, 255–262. [Google Scholar] [CrossRef]
  41. Nasal, A.; Sznitowska, M.; Buciński, A.; Kaliszan, R. Hydrophobicity Parameter from High-Performance Liquid Chromatography on an Immobilized Artificial Membrane Column and Its Relationship to Bioactivity. J. Chromatogr. A 1995, 692, 83–89. [Google Scholar] [CrossRef]
  42. Naseem, S.; Zushi, Y.; Nabi, D. Development and Evaluationof Two-Parameter Linear Free Energy Models for the Prediction of Human Skin Permeability Coefficient of Neutral Organic Chemicals. J. Cheminform. 2021, 13, 25. [Google Scholar] [CrossRef] [PubMed]
  43. Soriano-Meseguer, S.; Fuguet, E.; Port, A.; Rosés, M. Estimation of Skin Permeation by Liquid Chromatography. ADMET DMPK 2018, 6, 140–152. [Google Scholar] [CrossRef] [Green Version]
  44. Wang, Y.; Sun, J.; Liu, H.; Liu, J.; Zhang, L.; Liu, K.; He, Z. Predicting Skin Permeability Using Liposome Electrokinetic Chromatography. Analyst 2009, 134, 267–272. [Google Scholar] [CrossRef] [PubMed]
  45. Waters, L.J.; Shahzad, Y.; Stephenson, J. Modelling Skin Permeability with Micellar Liquid Chromatography. Eur. J. Pharm. Sci. 2013, 50, 335–340. [Google Scholar] [CrossRef]
  46. Sobańska, A.W.; Robertson, J.; Brzezińska, E. Application of RP-18 TLC Retention Data to the Prediction of the Transdermal Absorption of Drugs. Pharmaceuticals 2021, 14, 147. [Google Scholar] [CrossRef]
  47. Ciura, K.; Dziomba, S.; Nowakowska, J.; Markuszewski, M.J. Thin Layer Chromatography in Drug Discovery Process. J. Chromatogr. A 2017, 1520, 9–20. [Google Scholar] [CrossRef]
  48. Sobańska, A.W.; Brzezińska, E. IAM Chromatographic Models of Skin Permeation. Molecules 2022, 27, 1893. [Google Scholar] [CrossRef]
  49. Hong, H.; Wang, L.; Han, S.; Zou, G. The Estimation of Bioconcentration Factors of Aromatic Hydrocarbons by High-performance Liquid Chromatography. Toxicol. Environ. Chem. 1996, 56, 185–191. [Google Scholar] [CrossRef]
  50. Guo, R.; Liang, X.; Chen, J.; Zhang, Q.; Wu, W.; Kettrup, A. Using HPLC Retention Parameters to Estimate Fish Bioconcentration Factors of Organic Compounds. J. Liq. Chromatogr. Relat. Technol. 2004, 27, 1861–1873. [Google Scholar] [CrossRef]
  51. Sobanska, A. RP-18 TLC Retention Data and Calculated Physico-Chemical Parameters as Predictors of Soil-Water Partition and Bioconcentration of Organic Sunscreens. Chemosphere 2021, 279, 130527. [Google Scholar] [CrossRef]
  52. Tsopelas, F.; Stergiopoulos, C.; Tsakanika, L.-A.; Ochsenkühn-Petropoulou, M.; Tsantili-Kakoulidou, A. The Use of Immobilized Artificial Membrane Chromatography to Predict Bioconcentration of Pharmaceutical Compounds. Ecotoxicol. Environ. Saf. 2017, 139, 150–157. [Google Scholar] [CrossRef]
  53. Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-Likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep. 2017, 7, 42717. [Google Scholar] [CrossRef] [Green Version]
  54. U.S. Environmental Protection Agency. EPISuiteTM-Estimation Program Interface|USEPA. Available online: https://www.epa.gov/tsca-screening-tools/epi-suitetm-estimation-program-interface (accessed on 2 January 2023).
  55. Seung, L.J. EPISuite: A Fascinate Predictive Tool for Estimating the Fates of Organic Contaminants. J. Bioremediat. Biodegrad. 2016, 7, e171. [Google Scholar] [CrossRef] [Green Version]
  56. Stepanov, D.; Canipa, S.; Wolber, G. HuskinDB, a Database for Skin Permeation of Xenobiotics. Sci. Data 2020, 7, 426. [Google Scholar] [CrossRef]
  57. Lunghini, F.; Marcou, G.; Azam, P.; Patoux, R.; Enrici, M.H.; Bonachera, F.; Horvath, D.; Varnek, A. QSPR Models for Bioconcentration Factor (BCF): Are They Able to Predict Data of Industrial Interest? SAR QSAR Environ. Res. 2019, 30, 507–524. [Google Scholar] [CrossRef]
  58. Potts, R.O.; Guy, R.H. Predicting Skin Permeability. Pharm. Res. 1992, 9, 663–669. [Google Scholar] [CrossRef]
  59. Meylan, W.M.; Howard, P.H.; Boethling, R.S.; Aronson, D.; Printup, H.; Gouchie, S. Improved Method for Estimating Bioconcentration/Bioaccumulation Factor from Octanol/Water Partition Coefficient. Environ. Toxicol. Chem. 1999, 18, 664–672. [Google Scholar] [CrossRef]
  60. Fu, X.C.; Wang, G.P.; Wang, Y.F.; Liang, W.Q.; Yu, Q.S.; Chow, M.S.S. Limitation of Potts and Guy’s Model and a Predictive Algorithm for Skin Permeability Including the Effects of Hydrogen-Bond on Diffusivity. Pharmazie 2004, 59, 282–285. [Google Scholar]
  61. Cronin, M.T.D.; Dearden, J.C.; Moss, G.P.; Murray-Dickson, G. Investigation of the Mechanism of Flux across Human Skin In Vitro by Quantitative Structure–Permeability Relationships. Eur. J. Pharm. Sci. 1999, 7, 325–330. [Google Scholar] [CrossRef]
  62. Mitragotri, S.; Anissimov, Y.G.; Bunge, A.L.; Frasch, H.F.; Guy, R.H.; Hadgraft, J.; Kasting, G.B.; Lane, M.E.; Roberts, M.S. Mathematical Models of Skin Permeability: An Overview. Int. J. Pharm. 2011, 418, 115–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Equation (6)—predicted and experimental log Kp vs. reference values.
Figure 1. Equation (6)—predicted and experimental log Kp vs. reference values.
Pharmaceutics 15 01172 g001
Figure 2. Equation (7)—predicted and experimental log Kp vs. reference values.
Figure 2. Equation (7)—predicted and experimental log Kp vs. reference values.
Pharmaceutics 15 01172 g002
Figure 3. Equation (10)—predicted and experimental log BCF vs. reference values.
Figure 3. Equation (10)—predicted and experimental log BCF vs. reference values.
Pharmaceutics 15 01172 g003
Figure 4. Equation (11)—predicted and experimental log BCF vs. reference values.
Figure 4. Equation (11)—predicted and experimental log BCF vs. reference values.
Pharmaceutics 15 01172 g004
Table 1. Chromatographic retention factors and calculated physico-chemical properties of compounds 1 to 32.
Table 1. Chromatographic retention factors and calculated physico-chemical properties of compounds 1 to 32.
No.Compoundlog kKERlog kIAMMw#HvAt#ArHvAtFCsp3#FRB#HA#HDMRTPSA(N + O)log Kowlog S
12-Cresole−0.180.36108.1860.1401133.420.211.95−2.29
22-Naphtol 0.881.25144.21110001146.020.212.70−3.11
33-Cresole−0.220.36108.1860.1401133.420.211.96−2.30
43-Nitrophenol0.240.60139.1106013137.366.142.00−2.34
54-Bromophenol0.341.00173.086001136.220.212.59−3.10
64-Chlorophenol0.270.73128.686001133.520.212.39−2.70
74-Cresole −0.080.42108.1860.1401133.420.211.94−2.29
84-Ethylphenol−0.250.76122.2960.2511138.220.212.58−2.65
94-Nitrophenol0.190.60139.1106013137.366.141.91−2.28
10Baclofen−0.33−0.73213.71460.343255.363.33−0.96−0.61
11Chlorocresole0.681.18142.6960.1401138.420.212.70−3.09
12Methylparaben 0.040.52152.21160.1223139.746.531.96−2.29
13Phenol −0.270.3794.176001128.520.211.46−1.98
14Phenylalanine−0.20−0.65165.21260.2233245.563.33−1.44−0.08
15Resorcinol−0.38−0.14110.186002230.540.520.80−1.58
16Salcylic acid −0.06−0.58138.1106013235.457.532.26−2.50
17Thymol 0.521.34150.21160.411148.020.213.30−3.19
181,2,3-tris(1-methylethyl)benzene0.752.43204.41560.630070.20.006.36−4.54
191,4-dinitrobenzene0.450.16168.1126024044.191.661.46−2.04
203-(trifluoromethyl)phenol0.191.23162.11160.1414133.520.212.95−3.04
214-cyanophenol−0.050.77119.196002133.244.021.60−2.08
224-iodophenol0.801.59220.086001141.220.212.91−3.59
234-nitrobenzoic acid−0.23−0.23167.1126024142.283.151.89−2.30
24Anizole−0.090.31108.1860.1411032.99.212.11−2.33
25Benzamide−0.04−0.10121.196011134.543.120.64−1.42
26benzene −0.270.0978.166000026.40.002.13−2.41
27benzoic acid −0.21−0.74122.196012133.437.321.87−2.20
28Benzonitrile0.020.15103.186001031.223.811.56−2.02
29caffeine 0.08−0.40194.21490.3803052.061.86−0.07−1.48
30Chlorobenzene0.130.66112.676000031.50.002.84−2.96
31Indazole0.230.71118.199001136.128.721.77−2.72
32Toluene−0.050.4492.1760.1400031.40.002.73−2.77
Table 2. Reference (EPI), predicted, and experimental values of log Kp.
Table 2. Reference (EPI), predicted, and experimental values of log Kp.
log KpEPIEquation (6)Equation (7)Equation (8)Equation (9)log Kpexp
2-Cresole−5.58−5.71−5.67−5.89−5.57−5.36
2-Naphtol−5.26−4.99−5.00−5.05−5.26−4.76
3-Cresole−5.57−5.71−5.75−5.89−5.65−5.37
3-Nitrophenol−5.73−6.22−6.06−6.01−5.98−5.81
4-Bromophenol−5.52−5.20−5.74−5.30−5.74−5.00
4-Chlorophenol−5.39−5.42−5.18−5.55−5.09−5.00
4-Cresole−5.58−5.67−5.50−5.84−5.39−5.31
4-Ethylphenol−5.21−5.39−5.88−5.52−5.80−5.01
4-Nitrophenol−5.79−6.22−6.15−6.01−6.08−5.81
Baclofen−8.28−7.25−7.66−7.23−7.69
Chlorocresole−5.05−5.05−4.53−5.12−4.43−4.82
Methylparaben−5.84−5.98−6.09−5.94−6.02−5.63
Phenol−5.84−5.71−5.76−5.89−5.65−5.61
Resorcinol−6.40−6.43−6.48−6.52−6.40−6.63
Thymol −4.87−4.92−4.66−4.97−4.56−4.77
1,2,3-tris(1-methylethyl)benzene −3.78−3.73−4.13−3.80−4.06
1,4-dinitrobenzene−6.29−6.96−6.38−6.61−6.32
3-(trifluoromethyl)phenol −5.19−5.01−5.44−5.07−5.38
4-cyanophenol −5.89−5.74−5.96−5.68−5.86−5.73
4-iodophenol −5.58−4.71−5.64−4.74−5.70
Anizole−5.46−5.59−5.29−5.86−5.18
Benzamide −6.58−6.43−5.96−6.50−5.86
Benzene −5.26−5.63−5.22−6.00−5.09−4.51
Benzonitrile −5.82−5.94−5.31−6.12−5.19
Caffeine −7.53−6.96−7.38−6.91−7.66−7.56
Chlorobenzene −4.97−5.17−4.92−5.47−4.81
Indazole −5.44−5.56−5.94−5.63−6.11
Toluene −4.92−5.35−4.92−5.68−4.78−3.64
Table 3. Reference, predicted, and experimental values of log BCF.
Table 3. Reference, predicted, and experimental values of log BCF.
log BCFEPIEquation (10)Equation (11)Equation (12)Equation (13)log BCFexp
2-Cresole0.951.130.971.010.981.03
2-Naphtol1.451.471.481.431.45
3-Cresole0.961.130.941.010.941.23
3-Nitrophenol0.990.850.640.980.67
4-Bromophenol1.381.311.441.231.521.17
4-Chlorophenol1.241.141.291.031.371.42
4-Cresole0.951.171.041.061.06
4-Ethylphenol1.371.551.111.521.10
4-Nitrophenol0.930.850.600.980.630.71
Baclofen0.500.510.990.530.90
Chlorocresole1.451.641.771.631.90
Methylparaben0.961.080.931.120.94
Phenol0.630.920.710.760.71
Phenylalanine0.500.440.700.440.65
Resorcinol0.500.510.370.400.34
Salcylic acid 0.500.170.500.090.50
Thymol 1.842.132.032.232.121.48
1,2,3-tris(1-methylethyl)benzene3.863.203.413.403.49
1,4-dinitrobenzene0.630.460.620.670.65
3-(trifluoromethyl)phenol1.611.671.231.671.30
4-cyanophenol0.721.060.651.090.660.91
4-iodophenol1.591.671.961.682.10
4-nitrobenzoic acid 0.500.270.210.370.15
Anizole1.061.151.200.961.23
Benzamide0.500.530.720.430.73
Benzene1.070.840.980.531.00
Benzoic acid0.500.160.66−0.060.650.93
Benzonitrile0.700.770.960.601.00
Caffeine0.500.840.630.930.48
Chlorobenzene1.541.191.450.951.521.34
Indazole0.831.090.671.030.61
Toluene1.471.271.331.051.371.02
Table 4. Correlations ® between chromatographic and calculated parameters (n= 32).
Table 4. Correlations ® between chromatographic and calculated parameters (n= 32).
log kKERlog kIAMMw#HvAt#ArHvAtFCsp3#FRB#HA#HDMRTPSAlog Kowlog S
log kKER1.000.750.480.260.330.15−0.07−0.15−0.250.45−0.160.57−0.67
log kIAM0.751.000.200.000.060.26−0.19−0.42−0.310.26−0.540.81−0.85
Mw0.480.201.000.770.120.430.570.430.180.800.390.01−0.09
#HvAt0.260.000.771.000.250.620.750.560.130.890.54−0.080.11
#ArHvAt0.330.060.120.251.000.03−0.24−0.02−0.090.240.02−0.09−0.05
FCsp30.150.260.430.620.031.000.46−0.09−0.120.75−0.160.21−0.13
#FRB−0.07−0.190.570.75−0.240.461.000.490.250.660.48−0.180.30
#HA−0.15−0.420.430.56−0.02−0.090.491.000.380.180.87−0.450.45
#HD−0.25−0.310.180.13−0.09−0.120.250.381.000.000.37−0.420.40
MR0.450.260.800.890.240.750.660.180.001.000.230.16−0.14
TPSA−0.16−0.540.390.540.02−0.160.480.870.370.231.00−0.570.57
log Kow0.570.810.01−0.08−0.090.21−0.18−0.45−0.420.16−0.571.00−0.96
log S−0.67−0.85−0.090.11−0.05−0.130.300.450.40−0.140.57−0.961.00
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

Sobańska, A.W.; Brzezińska, E. Immobilized Keratin HPLC Stationary Phase—A Forgotten Model of Transdermal Absorption: To What Molecular and Biological Properties Is It Relevant? Pharmaceutics 2023, 15, 1172. https://doi.org/10.3390/pharmaceutics15041172

AMA Style

Sobańska AW, Brzezińska E. Immobilized Keratin HPLC Stationary Phase—A Forgotten Model of Transdermal Absorption: To What Molecular and Biological Properties Is It Relevant? Pharmaceutics. 2023; 15(4):1172. https://doi.org/10.3390/pharmaceutics15041172

Chicago/Turabian Style

Sobańska, Anna Weronika, and Elżbieta Brzezińska. 2023. "Immobilized Keratin HPLC Stationary Phase—A Forgotten Model of Transdermal Absorption: To What Molecular and Biological Properties Is It Relevant?" Pharmaceutics 15, no. 4: 1172. https://doi.org/10.3390/pharmaceutics15041172

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