Skip to main content
Erschienen in: Drugs in R&D 3/2023

Open Access 10.06.2023 | Original Research Article

Predictive Potential of Acido-Basic Properties, Solubility and Food on Bioequivalence Study Outcome: Analysis of 128 Studies

verfasst von: Dejan Krajcar, Rebeka Jereb, Igor Legen, Jerneja Opara, Iztok Grabnar

Erschienen in: Drugs in R&D | Ausgabe 3/2023

Abstract

Background and Objectives

Risk assessment related to bioequivalence study outcome is critical for effective planning from the early stage of drug product development. The objective of this research was to evaluate the associations between solubility and acido-basic parameters of an active pharmaceutical ingredient (API), study conditions and bioequivalence outcome.

Methods

We retrospectively analyzed 128 bioequivalence studies of immediate-release products with 26 different APIs. Bioequivalence study conditions and acido-basic/solubility characteristics of APIs were collected and their predictive potential on the study outcome was assessed using a set of univariate statistical analyses.

Results

There was no difference in bioequivalence rate between fasting and fed conditions. The highest proportion of non-bioequivalent studies was for weak acids (10/19 cases, 53%) and neutral APIs (23/95 cases, 24%). Lower non-bioequivalence occurrence was observed for weak bases (1/15 cases, 7%) and amphoteric APIs (0/16 cases, 0%). The median dose numbers at pH 1.2 and pH 3 were higher and the most basic acid dissociation constant (pKa) was lower in the non-bioequivalent group of studies. Additionally, APIs with low calculated effective permeability (cPeff) or low calculated lipophilicity (clogP) had lower non-bioequivalence occurrence. Results of the subgroup analysis of studies under fasting conditions were similar as for the whole dataset.

Conclusion

Our results indicate that acido-basic properties of API should be considered in bioequivalence risk assessment and reveal which physico-chemical parameters are most relevant for the development of bioequivalence risk assessment tools for immediate-release products.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s40268-023-00426-6.
Key Points
Acido-basic characteristics of active pharmaceutical ingredients (APIs) are associated with bioequivalence outcome.
APIs with lower calculated lipophilicity (clogP) are less likely to fail a bioequivalence study.
Bioequivalence studies in fasting and fed conditions have similar non-bioequivalence rates.

1 Introduction

Demonstration of safety and efficacy of a generic medicine by evaluating bioequivalence is always accompanied by certain risks. Whereas the patient risk (type I error) is appropriately controlled by the health authorities through validated testing methodology, there is less guidance on how industry can control the risk and avoid spending unnecessary time and resources on unsuccessful bioequivalence studies.
Bioequivalence study is usually one of the last steps in the development of a generic drug product. However, risk-based product development defined within a quality-by-design framework needs to start in the early planning phase. Early stage usually coincides with lack of data. Thus, bioequivalence risk assessment and mitigation strategy definitions initially rely on prior knowledge and understanding of how different interrelated factors impact bioequivalence study outcome.
The key method of classification impacting assessment of risk related to bioavailability is the Biopharmaceutics Classification System (BCS) [1], which is widely discussed within the literature. There are analyses available on real sets of data which assess BCS impact on bioequivalence outcome [25]. BCS is also implemented in assessments related to waiving of in vivo studies [6]. One of the extensions to BCS, the Biopharmaceutics Drug Disposition Classification System (BDDCS) [7], is a powerful tool for further insights into the mechanism of issues related to bioavailability. Nevertheless, it was not found to be advantageous for predicting bioequivalence study outcome, when compared with BCS [3].
We have previously presented results from a comprehensive study on a real set of bioequivalence study data and assessed the association between various pharmacokinetic parameters/active pharmaceutical ingredient (API) characteristics and bioequivalence study outcome [8]. Poorly soluble APIs resulted in non-bioequivalence study outcome significantly more often than highly soluble APIs. Poorly soluble group subsets of APIs (e.g., low bioavailability, P-glycoprotein substrates, first-pass metabolism, short time to peak plasma concentration, etc.) were found at increased risk for a conclusion of non-bioequivalence.
To improve bioequivalence risk assessment (i.e., to further differentiate the poorly soluble drug classes), one might investigate how poorly soluble the API really is (e.g., calculate dose number [Do] defined in BCS [9]) and/or classify APIs based on causes for poor intrinsic solubility. This can be assessed by general solubility equation (high partition coefficient [logP] and high melting point [Tm]) or, since a good proportion of APIs have ionizing functional groups, Henderson-Hasselbalch (H-H) equation (specific pKa). Several reports point out the relevance of acido-basic properties (estimated by pKa) in the new drug development [1012], bioequivalence assessment [13] and acceptability of bio-waivers and extension of BCS to subclasses considering pKa and aqueous solubility at pH 2 and pH 6.5 [14, 15]. Several publications differentiate the development of bio-relevant dissolution testing methodologies, in silico models and in vitro–in vivo correlations (IVIVC) based on weakly acid [14, 1620], neutral [21] or weakly basic properties of drugs [2224]. While these individual cases clearly indicate relevance of acido-basic properties in bioequivalence testing, there is, however, a lack of comprehensive research on a wider group of molecules available that would assess the association between acido-basic properties of APIs, intrinsic factors of solubility and bioequivalence study outcome under different study conditions.
Thus, the aim of this analysis was to extend our previous research [8] and explore the association between additional chemical/biopharmaceutical properties (solubility, logP, effective permeability, Tm, pKa and acido-basic properties) of poorly soluble APIs and their association with non-bioequivalence study outcome in fasting and fed conditions. This could help to further differentiate poorly soluble APIs with regards to bioequivalence outcome and clarify which API parameters are relevant for bioequivalence risk assessment.

2 Methods

2.1 Database Preparation

All data from in-house bioequivalence studies sponsored by Sandoz1 that satisfied the inclusion criteria were included in the analysis. Criteria for inclusion of the study into the database were as follows:
  • Bioequivalence study date of completion was within the pre-specified time period.
  • Study was a pivotal bioequivalence study (i.e., pilot studies were not in the scope of this analysis, since the power of such studies is very likely insufficient).
  • Post-hoc power was > 80% for primary pharmacokinetic parameters.
  • Study was completed (i.e., study phases were completed as per protocol and a report was issued).
  • Results were not inconclusive (i.e., there was no clinical or bioanalytical deviation that would impact study results).
  • Test and reference products were both immediate-release products.
  • Test and reference products contained one API or were fixed-dose combination products containing two or more APIs. In the latter case the study was treated as two or more independent bioequivalence studies for each API.
  • Bioavailability of test and reference products was compared under the same condition (either fasting or fed).
  • API was assessed as poorly soluble according to BCS.
Information collected or calculated for each study and/or API is presented in Table 1. Sandoz in-house or, where not available, literature solubility data were used. GastroPlus® v9.8 (Simulations Plus Inc., Lancaster, CA, USA) was used to retrieve calculated effective permeability (cPeff) and clogP for each API using the absorption, distribution, metabolism, excretion and toxicity (ADMET) Predictor® and .mol file of the API [25].cPeff was used to classify APIs into high and low cPeff group with a cut-off value of 3 for metoprolol (considered as a standard for high permeability). Zhou et al. concluded that when an API’s logP was < 3, the solubility of poorly soluble APIs was less impacted by (bio)surfactant variations [26]. Thus, our hypothesis was that when clogP is < 3 (low clogP), the risk for non-bioequivalent results will be lower compared with a high-clogP group, due to potentially lower variability of solubility limited absorption and/or lower impact of composition (surfactant) differences between compared treatments.
Table 1
Type of information collected or calculated for each bioequivalence study and/or each API
Parameter
Variable type
Unit of measurement
Description
BE outcome
Categorical (BE/non-BE)
No unit
Outcome of the study based on passing BE criteria for Cmaxa
Dose
Numerical
mg
Dose (D) of the API in the investigational product administered in the study
Food
Categorical (fasted/fed)
No unit
BE study is performed in fasted or fed conditions
Acido_basic_class
Numerical
No unit
Acido-basic classification based on dose number (see Table 2)
Solub_pH1.2b,c
Numerical
mg/mL
Equilibrium solubility at pH 1.2 and 37 °C (relevant pH for estimating solubility in fasted gastric condition)
Solub_pH3b,c
Numerical
mg/mL
Equilibrium solubility at pH 3 and 37 °C (relevant pH for estimating solubility in fed gastric conditions)
Solub_pH4.5b,c
Numerical
mg/mL
Equilibrium solubility at pH 4.5 and 37 °C (relevant pH for estimating solubility in fed gastric conditions)
Solub_pH6.8b,c
Numerical
mg/mL
Equilibrium solubility at pH 6.8 and 37 °C (relevant pH for estimating solubility in intestinal conditions)
Do_12
Numerical
No unit
Dose number at pH 1.2 (see Eq. 1)
Do_3
Numerical
No unit
Dose number at pH 3 (see Eq. 1)
Do_45
Numerical
No unit
Dose number at pH 4.5 (see Eq. 1)
Do_68
Numerical
No unit
Dose number at pH 6.8 (see Eq. 1)
clogP
Numerical
No unit
Calculated (predicted) partition coefficient (logP) determined by ADMET Predictor® of the GastroPlus® software
clogP_class
Categorical (low clogP/high clogP)
No unit
clogP < 3 was classified as low clogP and clogP ≥ 3 was classified as high clogP. Selection of cut-off value is explained in the methods section
Tm
Numerical
°C
Measured melting point [24]
pKa_ma
Numerical
No unit
Predicted pKa for the most acidic functional group—by Chemaxon ADMET [24]
pKa_mb
Numerical
No unit
Predicted pKa for the most basic functional group—by Chemaxon ADMET [24]
cPeff_class
Categorical (high cPeff/low cPeff)
cm/s ×10−4
Classification based on calculated human effective jejunal permeability determined by ADMET Predictor® of the GastroPlus® software with metoprolol cPeff as a cutoff value
ADMET absorption, distribution, metabolism, excretion and toxicity, API active pharmaceutical ingredient, AUC area under the plasma concentration–time curve, BE bioequivalence, clogP calculated partition coefficient, clogP_class ‘high’ or ‘low’ based on a cut-off clogP = 3, Cmax peak plasma concentration, cPeff calculated effective permeability, cPeff_class ‘high’ or ‘low’ based on a cut-off of metoprolol cPeff, D dose, Do dose number, logP partition coefficient, Peff effective permeability, pKa acid dissociation constant, pKa_ma predicted pKa for the most acidic functional group, pKa_mb predicted pKa for the most basic functional group, Tm melting point
aIn all cases, Cmax was more discriminating for conclusion of bioequivalence than AUC
bLiterature
cSandoz in-house data
Our database included bioequivalence studies under fasting and fed conditions. A bioequivalence study was considered successful when the bioequivalence criteria defined in the study protocol were satisfied and the compared products were concluded to have bioequivalence. In case of fixed-dose combination products, the success of bioequivalence study for each API was evaluated separately. Thus, the analysis was performed on 145 observations of 128 bioequivalence studies. For example, it could happen that for API 1 of a fixed-dose combination product, bioequivalence was concluded and for API 2, bioequivalence was not concluded. Such a product cannot be considered bioequivalent, but for the purpose of this analysis the outcome for API 1 was still considered as bioequivalent. The term non-bioequivalence is used intentionally since failure to conclude bioequivalence does not lead to conclusion of bioinequivalence.
Do for a specific pH was calculated with Equation 1:
$${\mathrm{Do}}_{\mathrm{pH}}=\frac{D \,\left(\mathrm{mg}\right)}{S\,\left(\mathrm{pH}\right) \times 250\, \mathrm{mL}},$$
(1)
where Do_pH is Do at a specific pH, D is the dose used in a bioequivalence study (mg) and S (pH) is solubility at specific pH (mg/mL).

2.2 Acido-Basic Classification

Drugs were classified as weak base, weak acid, amphoteric or neutral-like based on Do at gastrointestinal tract (GIT)-relevant pH (1.2, 3, 4.5 and 6.8). See Table 2 for classification criteria. If Do was ≤ 1, API was considered as highly soluble and if Do was ≤ 2 at any condition, the drug was considered borderline soluble under that condition. Any Do > 2 led to conclusion of low solubility. The initial idea for such classification came from the BCS subclasses proposed by Tsume et al. [15]. However, we have not differentiated between high and low permeability and introduced solubility at pH 4.5 into classification criteria. Namely, based on BCS-based biowaver guidelines, API is considered highly soluble if the highest dose is soluble in 250 mL of three buffers in a pH range of 1.2–6.8, preferably 1.2, 4.5 and 6.8 [6]. This leads to creation of an additional class not defined by Tsume et al., that is, amphoteric poorly soluble molecules. Note that many drugs classified as neutral-like also had acidic or basic functional groups. However, they were not ionizing in the GIT pH range or their ionization did not impact solubility.
Table 2
Conditions for classification of active pharmaceutical ingredients to different acido-basic groups based on dose numbers across gastrointestinal pH range
 
pH 1.2
pH 4.5
pH 6.8
Weak acid
Low solubility
Low solubility
High solubility OR borderline solubility
Weak base
High solubility OR borderline solubility
Low solubility
Low solubility
Amphoteric
High solubility OR borderline solubility
Low solubility
High solubility OR borderline solubility
Neutral-like
Low solubility
Low solubility
Low solubility
High solubility: Do ≤ 1; Borderline solubility: 1 < Do ≤ 2; Low solubility: Do > 2
We have also attempted to create acido-basic classification solely on the predicted pKa values. This classification of APIs to similar weak-acid, weak-base, amphoteric and neutral-like classes considered molecule structure, relevant GIT pH range (1.2–6.8) and pKa of the most acidic (pKa_ma) and the most basic (pKa_mb) functional groups of the API. With this information, proportion of ionization was calculated by H–H equation [27]. If < 1% of ionization was expected in the pH range, then the API was considered neutral. If ionization was > 1% anywhere in the GIT range (1.2–6.8), then, depending on the ionizable functional group(s), the APIs were classified as weak base, weak acid or amphoteric. For example, if a weak acid’s most acidic pKa was > 8.8, then this molecule was considered and classified as neutral in the GIT pH range, since < 1% would be ionized in the GIT pH range. For the same reason, amphoteric molecules were classified as weak bases if the most basic pKa of the drug was > 8.8.
When the two acido-basic classifications were compared, we noticed discrepancies, minor in classification of weak acids and weak bases and major in classification of neutral and amphoteric molecules. The reason could be in the error made by using predicted pKa. The solubility-based classification was considered more relevant since it was based on the measured solubility which plays a crucial role in the bioavailability of API. Thus, this solubility-based classification (as defined in Table 2) was used as a primary analysis.

2.3 Statistical Analysis

Descriptive statistical analysis was performed to summarize API characteristics. Descriptive statistics were calculated on a subset of poorly soluble APIs for the bioequivalent and non-bioequivalent group for comparison purposes. Descriptive statistics were reported as median and interquartile ranges since the distributions departed from normal (see Tables 3, 4).
Table 3
Ranges of numeric parameters related to active pharmaceutical ingredient (API) and bioequivalence studies for highly and poorly soluble API
Parameter
Range
clogP (no unit)
1.2 to 5.8
Do_12 (no unit)a
− 4.3 to 9.6
Do_3 (no unit)a
− 2.9 to 9.6
Do_45 (no unit)a
− 2.9 to 9.6
Do_68 (no unit)a
− 6 to 9.6
pKa_ma (no unit)
0.91 to 19.7
pKa_mb (no unit)
− 4.2 to 9.5
Tm (°C)
68 to 301
clogP calculated partition coefficient, Do_12 dose number at pH 1.2, Do_3 dose number at pH 3, Do_45 dose number at pH 4.5, Do_68 dose number at pH 6.8, pKa acid dissociation constant, pKa_ma predicted pKa for the most acidic functional group, pKa_mb predicted pKa for the most basic functional group, Tm melting point
aNatural log transformation
Table 4
Number of studies shown as variable (parameters) levels by bioequivalence outcome and descriptive statistic of parameters by bioequivalence outcome
Parameter
BE study outcome
p value
BE
Non-BE
Overall
111 (77)
34 (23)
 
Food
  
1a
 Fast
86 (76)
27 (24)
 
 Fed
25 (78)
7 (22)
 
Acido_basic class
  
0.001a
 Amphoteric
16 (100)
0 (0)
 Neutral
72 (76)
23 (24)
 
 Weak acid
9 (47)
10 (53)
 
 Weak base
14 (93)
1 (7)
 
cPeff_class
  
0.002a
 High cPeff
65 (69)
30 (31)
 
 Low cPeff
46 (92)
4 (8)
 
clogP_class
  
0.025a
 High clogP (≥ 3)
84 (72)
32 (28)
 
 Low clogP (< 3)
27 (93)
2 (7)
 
All BE (N = 145)
   
 Do_12$,*
1.8 (4.4)
3.6 (3)
0.004b
 Do_3$,*
2.2 (2.7)
3.9 (3.4)
0.016b
 Do_45$,*
3.2 (3.1)
3.8 (2.1)
0.603b
 Do_68$,*
2.2 (4.6)
3.4 (4.3)
0.585b
 cLogP$
3.7 (1.3)
4.2 (0.1)
0.032b
 Tm$
176 (44)
176 (17)
0.88b
 pKa_ma$
7.4 (6.2)
4.33 (5.2)
0.903b
 pKa_mb$
1.45 (8.6)
− 2.7 (2.5)
0.002b
BE fast conditions (N = 113)
   
 Do_12$,*
1.6 (4.4)
4.4 (3)
0.014b
 Do_3$,*
2 (3.2)
3.9 (2.6)
0.078b
 Do_45$,*
3 (3.1)
4 (2.7)
0.647b
 Do_68$,*
2.2 (4.1)
4.4 (4.1)
0.409b
 cLogP$
3.7 (1.5)
4.2 (0.1)
0.010b
 Tm$
177 (64)
176 (13)
0.649b
 pKa_ma$
7.4 (6.7)
4.3 (5.3)
0.785b
 pKa_mb$
1.5 (8.6)
−2.7 (4.5)
0.018b
BE fed conditions (N = 32)
   
 Do_12$,*
1.8 (3.5)
2.8 (2.1)
0.121b
 Do_3$,*
2.2 (2.7)
2.8 (1.8)
0.294b
 Do_45$,*
2.1 (3)
3.3 (2)
0.466b
 Do_68$,*
2.2 (5.1)
3.3 (4.1)
0.715b
 cLogP$
4.2 (1.6)
4.3 (0.1)
0.509b
 Tm$
176 (32)
176 (53)
0.600b
 pKa_ma$
7.4 (7)
8.2 (5.2)
0.732b
 pKa_mb$
−0.4 (7.8)
−3 (1.4)
0.017b
Data reported as number (%) of studies
BE bioequivalence, clogP calculated partition coefficient, clogP_class ‘high’ or ‘low’ based on a cut-off clogP = 3, cPeff calculated effective permeability, cPeff_class ‘high’ or ‘low’ based on a cut-off of metoprolol cPeff, Do_12 dose number at pH 1.2, Do_3 dose number at pH 3, Do_45 dose number at pH 4.5, Do_68 dose number at pH 6.8, pKa acid dissociation constant, pKa_ma predicted pKa for the most acidic functional group, pKa_mb predicted pKa for the most basic functional group, Peff effective permeability, Tm melting point
$Median (IQR)
*Natural log transformation
aFisher’s exact test
bKruskal–Wallis test
A set of univariate tests was performed to test how different variables are associated with the bioequivalence outcome. A Fisher’s exact test was utilized to test the association between categorical variables (Table 4) and bioequivalence outcome. Association between the numeric variables and bioequivalence outcome was assessed by non-parametric Kruskal–Wallis tests due to deviations from the normal distribution. p Values of < 0.1 and < 0.05 were set for conclusion of association and strong association, respectively. As this was an exploratory analysis there was no correction for multiple comparisons. Tests used for specific parameters are presented in Table 4.
To assess the association between numeric parameters, non-parametric Spearman rank correlation tests were performed and Spearman coefficients (and associated p values) were used to assess the correlation. Correlation was assessed as weak, moderate and strong for absolute values of Spearman coefficients < 0.5, 0.5–0.75 and > 0.75, respectively.
To assess how well each significant parameter alone distinguishes between bioequivalence and non-bioequivalence outcome, we have created receiver operating characteristics (ROC) curves, calculated area under the ROC curve (AUROC) and its 95% confidence interval limits to assess if AUROC is significantly different from 0.5 (better than random guess). Youden index analysis was performed to find the optimal cutoff value (Youden threshold) providing the best tradeoff between sensitivity and specificity. The specificity and sensitivity at this threshold were also reported.
Data were analyzed using Minitab® v19.2020.3 (Minitab, Inc., State College, PA, USA) and R software v4.1.2 (R Core Team 2021, Vienna, Austria) using packages pROC v1.16.2 and corrplot v0.92.

3 Results

Ranges for numeric variables are summarized in Table 3. Results of association tests between different variables and bioequivalence outcome are summarized in Table 4.
No association was found within poorly soluble APIs between fasting or fed conditions and bioequivalence outcome of the study. Fasted and fed studies were failing at similar rates, 24% and 22%, respectively. There is no case in the pool of 128 studies where results under fed conditions would be more discriminatory than the results under fasting conditions, that is, no case where a study under fed conditions would be non-bioequivalent and a study under fasting conditions would be bioequivalent for the same molecule.
Strong association was shown between bioequivalence outcome and acido-basic properties. The riskiest were weak acids and neutral APIs with 53% and 24% non-bioequivalence rate, respectively. Weak bases and amphoteric APIs had 1 (7%) and 0 occurrence of non-bioequivalence, respectively.
Strong association was found between bioequivalence outcome and cPeff. We observed an 8% non-bioequivalence rate for APIs with low permeability (low cPeff) and a 31% non-bioequivalence rate for APIs with high permeability (high cPeff); 90%, 67%, 0% and 100% of weak-acid, neutral, amphoteric and weak-base APIs, respectively, were classified into high cPeff class.
Strong association was found between bioequivalence outcome and clogP_class. Our hypothesis was confirmed, since we observed a non-bioequivalence rate of only 7% when clogP was low, compared with 28% when clogP_class was high; 100%, 84%, 56% and 53% of weak-acid, neutral, amphoteric and weak-base APIs, respectively, were classified into high clogP class.
Analysis regarding the association of acido-basic properties, clogP_class and bioequivalence outcome was repeated on the fasting and fed subgroups (results not shown); under fasting conditions the conclusions were the same as on the complete dataset, whereas the fed subgroup was too small for any relevant conclusions.
Natural logarithms of Do at pH 1.2 (Do_12) and pH 3 (Do_3) were found to be significantly higher in the non-bioequivalent group. This was similar in the sub-analysis of studies under fasting conditions. However, the significance was lost in the fed subgroup analysis. Similarly, clogP values were significantly higher for the non-bioequivalent group, but the significance was lost in the fed subgroup analysis. pKa of the most basic functional group (pKa_mb) was found to be significantly lower for the non-bioequivalent group in the analysis on the complete set of data, as well as in the fasting and fed subgroup analyses.
AUROC values for above-mentioned significant parameters Acido_bacic_class, cPeff_class, clogP_class, Do_12, Do_3, clogP and pKa_mb were 0.69, 0.65, 0.59, 0.66, 0.63, 0.64 and 0.68, respectively (Fig. S1 in the electronic supplementary material [ESM]) and were all significantly different from 0.5.
Apart from these, no other API parameter was found to differ between non-bioequivalent and bioequivalent groups.
Strong correlation was found between the following pairs of dose numbers: Do_12 and Do_3; Do_45 and Do_68. All other correlations were moderate or weak (Fig. S2 in the ESM).

4 Discussion

4.1 Fasting Conditions

The highest incidence of non-bioequivalence outcome (53% non-bioequivalence rate) in the weak-acid group correlates with higher Do_12, Do_3 and significantly lower pKa value for the most basic functional group of the non-bioequivalent API group. In an attempt to explain this higher incidence of non-bioequivalence, it is important to note that 90% of these weak acids were also highly permeable (had cPeff higher than high permeability standard metoprolol and all had high clogP [≥ 3]) and would be classified as Class II-a according to the extended BCS classification [15]. These APIs are practically insoluble in gastric pH (without the solubility or dissolution enhancing excipients) and are subjected to highly variable gastric emptying. When they do reach the intestine, the conditions for solubility/dissolution are favorable due to ionization of the weak-acid APIs in the basic environment. Since these APIs are also highly permeable (due to passive or transporter-mediated absorption [2831]), they are readily absorbed, which means that the dissolution rate and potential formulation differences directly influence bioavailability [9]. This can explain the higher non-bioequivalence rate but at the same time points to the risk mitigation strategy. Namely, highly permeable weak-acid APIs are good candidates for building a Level A IVIVC, which was also the reason a BCS-based biowaiver extension was proposed for BCS Class II-a APIs [32]. Case-by-case research is needed to find biorelevant media to support such biowaivers [14]. There are several published successful product development cases and biowaiver applications of Level A IVIVC for weak-acid APIs [1618, 20].
The group of molecules with the second highest non-bioequivalence occurrence (24%) were neutral molecules, which contributed to higher median clogP values for the non-bioequivalent group. Nevertheless, 16% of the neutral molecules also had clogP < 3, which could have contributed to the lower non-bioequivalence occurrence compared with the weak-acid group. As hypothesized, solubility (and potentially absorption) of neutral APIs with lower clogP could be less impacted by variations in (bio)surfactants. In a related aspect, this group of neutral APIs included two thirds of highly permeable and one third of poorly permeable APIs and cPeff was significantly associated with the bioequivalence outcome. This cPeff difference may again help us in interpreting the results. Similar to a weak acid, if the permeability of a neutral API is high, the solubility/dissolution rate (and potential differences in formulations) would directly reflect the extent and rate of bioavailability. If the permeability is low, it limits the absorption and the potential difference in formulations might not impact the extent or rate of bioavailability. Thus, presence of poorly permeable neutral APIs (BCS subclass IVc) and APIs with clogP < 3 may contribute to a lower non-bioequivalence rate when compared with the weak-acid API group. Regardless, the 24% risk for non-bioequivalence needs to be mitigated during development, and examples of IVIVC for riskier BCS subclass IIc drugs include introduction of a biphasic system (absorptive compartment) into a gastrointestinal simulator and United States Pharmacopeia (USP) dissolution apparatus II [21].
It is worth adding that APIs in our database classified as weak acid or neutral were in larger proportion subject to first-pass metabolism and P-gp mediated efflux compared with weak bases and amphoteric molecules, which could also contribute to challenges with bioavailability [7] and bioequivalence of the neutral and weak-acid group of poorly soluble APIs.
In a weak-base API group, we could observe only one non-bioequivalent result. This phenomenon could again be explained by the fact that all APIs in a weak-base group had high permeability (cPeff more than those of metoprotol) and would be classified as BCS Class IIb APIs. Unlike weak acids, weak bases are completely ionized at gastric pH and they have a high potential to be completely dissolved (although incomplete dissolution may still occur [24]) from immediate-release formulations. However, when reaching high intestinal pH, there is a potential for precipitation, which can impact the absorption processes [33]. There is an alternative scenario where the API is retained in a supersaturated state (or with minimal precipitation) when passing through intestines. This could be retained by the presence of bile salts, excipients and/or fast absorption of the API from the intestine—and the higher the permeability, the higher the absorption rate of the API. Our observations support the hypothesis that highly permeable weak bases pose fewer challenges for bioequivalence due to lower probability of the precipitation phenomenon. On a separate note, there are successful IVIVC approaches available for BCS Class IIb APIs which could decrease the risk for non-bioequivalent results. These entail specific in vitro methods (e.g., using a multi-compartment GIT simulator) and in silico models for assessing complex dissolution, supersaturation and precipitation mechanisms [2224, 34] and are encouraged for use when developing products with weak-base APIs.
Another reason for observing low occurrence of non-bioequivalent results in the weak-base API group may be the smaller sample size.
The last group of poorly soluble APIs were amphoteric. There were no non-bioequivalent results for these APIs in the 13 fasting bioequivalence studies. These APIs had high solubility at gastric and intestinal pH. As such, they would be dissolved in the stomach and stay dissolved when reaching the intestine. However, when assessing their potential for a BCS biowaiver they would be considered as poorly soluble APIs (due to poor solubility at pH 3 or 4.5), even though they would resemble highly soluble APIs under fasting conditions. Thus, they could potentially be considered for BCS biowaiver approaches. Based on our results, the required testing of solubility across the whole pH range to classify an API as highly soluble for a BCS biowaiver may be too strict. Although we have not seen any non-bioequivalence in three fed bioequivalence studies, more research is needed to assess the risk there, since fed gastric conditions may increase pH to where these amphoteric APIs would behave like poorly soluble weak acids. However, it seems that even in this case the risk is lower, since gastric emptying may be the rate-limiting step for absorption in fed conditions, not the dissolution rate.

4.2 Fed Conditions

Our analysis showed no association between non-bioequivalence outcome and fasting/fed conditions (see Table 4). The 2% difference between percent of non-bioequivalence outcome within fasting and fed bioequivalence studies was far less than the approximately 15% difference observed by Tanguay et al. [4]. They reported 10–12% non-bioequivalence studies in fed conditions and 26.7% non-bioequivalence studies under fasted conditions (N = 1200 bioequivalence studies). The difference we observed is, however, closer to that observed by Lamouche et al., who reported a 14% failure rate within fasted bioequivalence studies and a 10% failure rate within fed bioequivalence studies within BCS Class I APIs (N = 918 bioequivalence studies) [5].
The fed subgroup analyses showed loss of significant differences for Do_12, cPeff and clogP between bioequivalent and non-bioequivalent groups, but the difference remained for the most basic pKa. The loss of significance can be caused by reduced power due to the lower sample size of the fed subgroup. However, it can also be explained by different GIT conditions caused by ingestion of a meal. There was less difference in median Do_12 for the bioequivalent and non-bioequivalent groups in fed compared with fasting conditions. Namely, the pH in the stomach increases to around 3 (or 3.5) in fed conditions [35] and the API solubility at pH 1.2 could become less relevant for prediction of in vivo behavior. But surprisingly, Do_3 and Do_45 also do not differ significantly between non-bioequivalence and bioequivalence in the fed condition, as well as in fasting. On the other hand, there was almost no difference in median clogP between the bioequivalent and non-bioequivalent groups in fed conditions, whereas it was significant in fasting. The ingestion of a meal triggers higher secretion of solubilizing bile salts, prolongs gastric emptying time and increases GIT fluid volumes, etc. [35] and all these effects could make solubility (e.g., Do) and lipophilicity (e.g., clogP) related parameters less relevant for assessing bioequivalence outcome in fed conditions. Interestingly, what remains unexplained is the significant difference of most basic pKa between the non-bioequivalent and bioequivalent groups, where the absence of basic functional groups in APIs (weak acids) still seems important for bioequivalence outcome even after meal ingestion, but apparently not by impacting solubility.
Fed bioequivalence studies in our database never had more discriminatory power to conclude non-bioequivalent results compared with fasted studies, that is, they always concluded the same outcome as studies in fasted conditions or passed bioequivalence when fasting study concluded non-bioequivalent results. This is acknowledged by most regulatory agencies, that is, that studies in fasted conditions are more discriminatory to detect differences between the test and reference product. Some agencies still request bioequivalence in fasted and fed conditions when a medicinal product is to be taken regardless of food and the product is not considered complex (e.g., solid dispersion). To avoid potentially non-discriminatory in vivo fed studies, alternatives are already suggested in the literature (e.g., PBPK modeling [36]).

5 Conclusion

Analyzing the database of 128 bioequivalence studies with 26 poorly soluble APIs, we found no difference in non-bioequivalence rate in fasting and fed conditions. Fed bioequivalence study conditions were never more discriminatory than fasting conditions.
Acido-basic characteristics were, however, associated with the bioequivalence outcome. The highest non-bioequivalence proportions were among weak acids (10/19 cases, 53%) and neutral API groups (23/95 cases, 24%). Lower non-bioequivalence occurrence was observed among weak bases (1/15 cases, 7%) and amphoteric APIs (0/16 cases, 0%). These findings were supported by higher median Do_1.2 and Do_3 and lower most basic pKa of the non-bioequivalent groups. Amphoteric poorly soluble APIs were shown as less risky. We discussed how differences between weak-acid, neutral and weak-base APIs may be somewhat confounded with differences in lipophilicity (clogP) or permeability (cPeff) of these APIs. Namely, high clogP and high cPeff classes were both associated with higher non-bioequivalence occurrence.
We pointed out a different example of risk mitigation strategies within each acido-basic group and indicated that amphoteric APIs might be relevant candidates for BCS-based biowaiver.
Subgroup analysis under fasting conditions had similar conclusions as the analysis on the whole dataset, whereas in the fed subgroup analysis the significance was lost for most parameters (with the exception of the most basic pKa). This finding could be attributed to numerous changes in gastrointestinal conditions after ingestion of a meal.
With these conclusions we have demonstrated the importance of acido-basic properties and added another piece of the puzzle in understanding the bioequivalence risk of the product at a very early stage of product development. There are limitations to our work that include the univariate analyses approach, especially since some of the parameters are correlated. The AUROC values between 0.6 and 0.7 can be labeled as satisfactory, but also indicate that the only path forward is a multivariate analysis. Different tools are available for addressing these issues, from multiple linear or logistic regression to more complex methods (e.g., machine learning techniques).
The limitations of our work can also be attributed to the in silico estimated parameters (clogP, cPeff, pKa), but at the same time, this limitation is also an advantage since these parameters are easily acquired at an early stage of product development and based on our results have predictive value for bioequivalence outcome. In addition, we only deal with API solubility in our research, however, for improved assessment, solubility of the intermediate (granulate, solid dispersion, etc.) might be more appropriate to assess the risk of bioequivalence. Knowledge about composition and process differences along with comparative in vitro dissolution profiles and IVIVC can certainly increase understanding of the risk prior to bioequivalence study, but this was not the purpose of our research.
One should use findings presented in this manuscript as a basis for further development of early-stage bioequivalence risk assessment methodology.

Acknowledgements

The authors would like to acknowledge coworkers at Sandoz for conducting trials, sharing data and valuable consultations for interpretation of data.

Declarations

Funding statement

Iztok Grabnar acknowledges support from the Slovenian Research Agency, research core funding no. P1-0189. This research did not receive any other specific grant from funding agencies in the public, commercial, or non-for-profit sectors.

Conflict of interest

Dejan Krajcar, Rebeka Jereb, Igor Legen and Jerneja Opara declare employment at Sandoz d.d. Iztok Grabnar is employed at University of Ljubljana, Faculty of Pharmacy. The authors declare that they have no conflicts of interest.

Author contribution (CreDIT)

DK: conceptualization, data curation, writing—original draft, methodology, formal analysis, visualization. RJ: data curation, writing—review and editing. IL: conceptualization, supervision, writing—review and editing. JO: writing—review and editing. IG: writing—review and editing.

Ethical considerations

All in vivo human bioequivalence studies that were included in this research were reviewed and approved for conduct by formally constituted review boards (Institutional Review Board or Ethics committee). All clinical trials included in this research were conducted in accordance with 1964 Declaration of Helsinki and its latest amendments, good clinical practice guidelines and other local regulatory laws and guidelines and were approved by the applicable Ethics Committee.
Written informed consent was obtained from all participants in all clinical trials included in this research.
Not applicable.

Code availability

Not applicable.

Availability of data and materials

Not applicable.
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.
Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Fußnoten
1
Lek Pharmaceuticals d.d., A Sandoz Company, Verovskova 57, 1526 Ljubljana, Slovenia.
 
Literatur
3.
Zurück zum Zitat Cristofoletti R, Chiann C, Dressman JB, Storpirtis S. A comparative analysis of biopharmaceutics classification system and biopharmaceutics drug disposition classification system: a cross-sectional survey with 500 bioequivalence studies. J Pharm Sci. 2013;102(9):3136–44. https://doi.org/10.1002/jps.23515.CrossRefPubMed Cristofoletti R, Chiann C, Dressman JB, Storpirtis S. A comparative analysis of biopharmaceutics classification system and biopharmaceutics drug disposition classification system: a cross-sectional survey with 500 bioequivalence studies. J Pharm Sci. 2013;102(9):3136–44. https://​doi.​org/​10.​1002/​jps.​23515.CrossRefPubMed
4.
Zurück zum Zitat Tanguay M, Potvin D, Haddad J, Lavigne J, et al. When will a drug formulation pass or fail bioequivalence criteria? Experience from 1200 studies. AAPS PharmSciTech. 2002;4(4):Abstract R6193. Tanguay M, Potvin D, Haddad J, Lavigne J, et al. When will a drug formulation pass or fail bioequivalence criteria? Experience from 1200 studies. AAPS PharmSciTech. 2002;4(4):Abstract R6193.
5.
Zurück zum Zitat Lamouche S, Leonard H, Shink É, Tanguay M. The biopharmaceutical classification system: can it help predict bioequivalence outcome? A CRO retrospective analysis. AAPS J. 2008;10(S2). Lamouche S, Leonard H, Shink É, Tanguay M. The biopharmaceutical classification system: can it help predict bioequivalence outcome? A CRO retrospective analysis. AAPS J. 2008;10(S2).
11.
Zurück zum Zitat Manallack DT. The pK(a) distribution of drugs: application to drug discovery. Perspect Med Chem. 2007;1:25–38. Manallack DT. The pK(a) distribution of drugs: application to drug discovery. Perspect Med Chem. 2007;1:25–38.
32.
Zurück zum Zitat Khalid F, Hassan SMF, Noor R, Zaheer K, Hassan F. Possibility of extending biopharmaceutics classification system based biowaiver to BCS class IIa drug. Pak J Pharm Sci. 2019;32(5):2065–73.PubMed Khalid F, Hassan SMF, Noor R, Zaheer K, Hassan F. Possibility of extending biopharmaceutics classification system based biowaiver to BCS class IIa drug. Pak J Pharm Sci. 2019;32(5):2065–73.PubMed
Metadaten
Titel
Predictive Potential of Acido-Basic Properties, Solubility and Food on Bioequivalence Study Outcome: Analysis of 128 Studies
verfasst von
Dejan Krajcar
Rebeka Jereb
Igor Legen
Jerneja Opara
Iztok Grabnar
Publikationsdatum
10.06.2023
Verlag
Springer International Publishing
Erschienen in
Drugs in R&D / Ausgabe 3/2023
Print ISSN: 1174-5886
Elektronische ISSN: 1179-6901
DOI
https://doi.org/10.1007/s40268-023-00426-6

Weitere Artikel der Ausgabe 3/2023

Drugs in R&D 3/2023 Zur Ausgabe