Bcs Classification Database

Posted by admin

Our resources page contains links to presentations, posters, abstracts, and links to the Drug Delivery Foundation's BCS database. Presentations We have active research programs related to preclinical technology transfer, oral drug delivery and anti-infectives therapeutics, including links to selected publications, presentations, and downloads.

  1. Bcs Classification System Database
  2. Bcs Classification Database Fda
  3. Bcs Classification Guidance
Published online 2015 Apr 29. doi: 10.1208/s12248-015-9743-7
PMID: 25916691
This article has been cited by other articles in PMC.

Abstract

The Biopharmaceutical Classification System (BCS) has been a prognostic tool for assessing the potential effects of formulation on the human drug oral bioavailability. When used in conjunction with in vitro dissolution tests, the BCS can support the prediction of in vivo product performance and the development of mechanistic models that support formulation assessments through the generation of “what if” scenarios. To date, the applicability of existing human BCS criteria has not been evaluated in dogs, thereby limiting its use in canine drug development. Therefore, we examined 50 drugs for which absolute bioavailability (F) was available both in dogs and humans. The drugs were also evaluated for any potential association between solubility (calculated from the dose number, Do) or lipophilicity (LogP) and F in dogs. In humans, solubility is determined in 250 mL of fluid. However, the appropriate volume for classifying drug solubility in dogs has not been established. In this analysis, the estimated volume of a water flush administered to fasted dogs (6 mL) and a volume of 250 mL scaled to a Beagle dog (35 mL) were examined. In addition, in humans, a Do value greater than 1.0 is used to define a compound as highly soluble and a LogP value greater than 1.72 as high permeability. These same criteria were applied for defining highly soluble and highly permeable in dogs. Whether using 35 or 6 mL to determine Do, the canine solubility classification remained unchanged for all but seven compounds. There were no clear associations between a drug’s F in dogs and humans or between the canine value of F and either its human BCS classification, its LogP value, or the canine Do estimate. There was a tendency for those drugs with canine values of F equal to or greater than 80% to have LogP values equal to or greater than 1.0. Exceptions to this observation tended to be those compounds known to be absorbed via mechanisms other than passive diffusion (e.g., via transporters or paracellular transporters). Although there are limitations to the approach used in this study, the results of our assessment strongly suggest that the human BCS classification system requires substantial modification before it can be reliably applied to dogs.

KEY WORDS: BCS, dogs, canine, permeability, solubility

INTRODUCTION

The United States Pharmacopeial Convention (USP) authorized the creation of an advisory panel to investigate the possibility of applying the principles of the Biopharmaceutics Classification System (BCS) to veterinary drugs—specifically, solid oral formulations administered to dogs (1). Developed for human pharmaceutical compounds (–), the BCS is an important tool that facilitates product development and regulatory decisions. By understanding the solubility of a compound in biorelevant media and its permeability across biological membranes, the rate limiting factors determining the rate and extent of oral drug absorption can be identified. This information can be invaluable for predicting the potential influence of formulation and physiological variables on oral drug bioavailability.

Within the framework of human pharmaceuticals, drugs can be classified into one of the following four BCS categories:

  • Class I: high solubility, high permeability: generally very well-absorbed compounds

  • Class II: low solubility, high permeability: exhibits dissolution rate-limited absorption

  • Class III: high solubility, low permeability: exhibits permeability-limited absorption

  • Class IV: low solubility, low permeability: very poor oral bioavailability

A complementary classification system was proposed by Wu and Benet (, ). They recognized that drugs exhibiting high permeability are generally extensively metabolized, while poorly permeable compounds are primarily eliminated as unchanged drug in the bile and urine. Thus, the Biopharmaceutical Drug Disposition Classification System (BDDCS) has been used to predict drug disposition and potential drug-drug interactions in the intestine and the liver and potentially the kidney and brain. Although the solubility criteria for the BCS and BDDCS are the same, there is a substantial difference in the second variable being considered. For the BDDCS, the second classification is related to the extent of drug metabolism. Conversely, the assessment of permeability in the BCS is linked to the extent of intestinal absorption, i.e., a drug is considered to be highly permeable when the extent of the systemic absorption (parent drug plus metabolites) in humans is determined to be at least 90% of an administered dose based on a mass balance determination or in comparison to an intravenous reference dose. Accordingly, the BCS and BDDCS classification of a drug may differ.

The US Food and Drug Administration’s (FDA’s) Center for Drug Evaluation and Research (CDER) (9) has incorporated BCS concepts into guidance documents for human medications into the 2000 FDA Guidance for Industry, including guidance for the waiver of in vivo bioequivalence study requirements for high solubility/high permeability drug products. However, the BCS has not as yet been extrapolated for application to veterinary drugs. The reason for this gap is that the BCS was developed based upon human digestive physiology, which can be vastly different from that observed in veterinary species. Given the similarity of therapeutic entities used in the dogs and humans, and because of the use of the dog as a preclinical species for human medicine (), it would be of particular value to have an understanding of how the BCS criteria can be translated between human and canine gastrointestinal (GI) physiologies.

The solubility criteria used both by the BCS and the BDDCS rely upon formulation considerations in that it is based upon the highest dose that will be administered. Both the BCS and the BDDCS define a high solubility compound at the highest marketed dose strength that is soluble in 250 mL of water over the pH range of 1–7.5 at 37°C. This definition differs from that of “intrinsic solubility,” which reflects the equilibrium aqueous solubility of a compound. For acids and bases, intrinsic solubility represents the concentration of the unionized species in a saturated solution at the pH value where that compound is fully unionized (). While there has been some debate regarding certain compounds whose intrinsic solubility may not be accurately defined when using conventional media (, 13), those considerations are founded upon the perspective of a drug’s physicochemical characteristics rather than on the in vivo conditions into which that drug will be introduced. Thus, while intrinsic solubility is solely a function of the molecule, the BCS (or BDDCS)-based solubility criteria is dependent upon physiological conditions and the corresponding targeted therapeutic dose. Unfortunately, what constitutes the BCS-based criteria for high or low solubility is currently undefined for dogs because of complexities associated with interspecies differences in the composition of the GI milieu (1).

Another obstacle confronted when trying to establish canine-specific BCS criteria is the challenge associated with the classification of intestinal permeability. Despite the range of high throughput systems available for examining human intestinal permeability, such as Caco-2 cells, parallel artificial membrane permeability assay (PAMPA), and phospholipid vesicle-based permeation assay (PVPA), these methods for estimating drug permeability have only been applied to human drugs (, ). These systems have not been developed and validated for application to drug permeability across the canine intestine (, ). Moreover, while one may argue that transcellular permeability should be similar in humans and dogs, the GI tract of the dog tends to be more permeable (leakier) because of the larger intercellular pores ().

Currently, the existing in vitro methods for evaluating drug permeability have not succeeded in providing data that can be extrapolated to dogs. For this reason, we needed to resort to comparisons based upon the use of absolute bioavailability. Because there are no suitable in vitro methods to assess effective permeability in dogs (Peff), we have used absolute bioavailability (F) for this analysis. We have justified this approach because a comparison of drug absorption across human colonic epithelium cell layers (Caco-2 cell line) to absorption across canine colon tissue did not show a relationship (). The Ussing chamber technique, which has been evaluated for other veterinary species (), has not been applicable for canine studies () because of the fragility of the tissue. Membrane damage that occurs prevents permeability measurements using this technique in dogs (). Therefore, without the availability of these in vitro tools, other data must be used to predict permeability and apply BCS criteria for oral drugs administered to dogs.

The current investigation was undertaken because of the lack of established BCS criteria to evaluate oral medications administered to dogs. The objectives were to examine the properties that define human BCS criteria for drugs and to compare this information to pharmacokinetic data available from studies in dogs. Without in vitro intestinal permeability data in dogs, another parameter must be considered to classify a drug as either high or low permeability. To this end, there are numerous molecular factors that impact drug transcellular permeability, including hydrogen bonding properties, molecular size and shape, polarity, flexibility, and ionization properties (, ). There is no “gold standard” or [even a suggested criterion based upon the log of the lipophilicity coefficient (LogP) pH-dependent lipophilicity value (LogD)] that has been proposed as a cutoff value. Therefore, we focused on the use of the systemic absorption value (absolute bioavailability, F), which we were able to obtain from the published literature. It was assumed that if the value of F is high, permeability (via active or passive processes) must likewise be high. We also acknowledged at the outset that the converse was not necessarily true and that the use of F as an indicator of drug permeability will produce some false negative results, i.e., there are drugs that have high permeability, but low values of F because of other factors such as intestinal efflux or high first-pass metabolism.

A drug solubility classification has not been established for medications administered to dogs (, 25). To classify a drug as either high or low solubility in dogs requires that one knows the ideal volume in which to measure solubility. In this study, we have examined two different volumes and considered whether this parameter can be useful to predict oral absorption of medications in dogs.

Ultimately, the objective of this study was to identify the in vitro drug properties with respect to their potential impact on dog-human differences and similarities on oral drug solubility and permeability. The foundational assumption was that if properties can be identified, we could then generate dog-specific criteria for applying BCS concepts to understand the critical formulation and physiological variables that can influence canine oral drug absorption. Similar to its tremendous influence on human drug product development and regulatory evaluation, a roadmap for screening oral product formulations, if applied to veterinary drug products, would provide a tool for screening new formulations. Additional benefits that would be associated with a canine-specific BCS would be an improvement on our ability to compare human formulations for potential testing and clinical use based upon information obtained in dogs (and vice versa).

One of our objectives was to extend our assessment beyond the results reported by Chiou et al., () in which the oral bioavailability of 43 compounds was compared in humans and dogs. In that study, they observed that while 22 of the 43 compounds were completely absorbed in both humans and dogs, the overall interspecies correlation of F values was low (coefficient of determination, R2 = 0.51). A pitfall associated with the investigation by Chiou et al. was that much of the data were based upon radiolabeled data, thereby precluding a differentiation between parent compound and metabolites. Given the potential for interspecies differences in intestinal metabolism, and since F values were based on total urinary recovery of radioactivity of the drug (thereby further confounding the comparison with potential differences in post-absorption processes), we could not use that information to generate predictions on the permeability component of the BCS. However, we also recognized that a drug with a high first-pass effect may be reported with high F in the study by Chiou et al. (), but not in our study reported. Thus, in addition to evaluation of BCS classification versus F in dogs and humans, we compared our F values with those reported by Chiou et al. ().

EXPERIMENTAL METHODS

To explore the potential application of BCS principles for oral drugs administered to dogs, pharmacokinetic, lipophilicity, and solubility data were either calculated or were obtained from existing literature. Lipophilicity, calculated as the octanol / water partition coefficient (LogP) was used to estimate permeability because there is no in vitro permeability data available for dogs, we used the experimental LogP values (which consider the ratio of unionized solute concentrations in octanol versus that in water), rather than Log D (which is ratio of the concentration of drug in octanol versus the sum of the concentration of both the neutral and ionized moieties in water) primarily because we found the LogP values to be more readily available for all 50 compounds in our study and because LogP had previously been included as a surrogate for predicting the intestinal permeability for human drugs (–). The list of drugs for which human BCS criteria were already derived was used as a starting database (–). Values for current BCS classification, solubility (expressed as mg/mL), and LogP were obtained from an internet web site that lists these criteria (Therapeutic Systems Research Laboratories (TSRL), Inc; http://tsrlinc.com/resources/services/) (27). Solubility data (mg/mL) and LogP estimates were also obtained from the literature (–). From the hundreds of drugs listed in the references cited above, the literature was searched for data on oral absorption of those compounds for which we were able to obtain data in dogs. The F values for dogs were obtained from published sources, the manufacturer’s data (approved drug label), or in some instances from the author’s unpublished research. Although we found additional published information on the oral bioavailability for some veterinary drugs (), human data and human BCS criteria were not available for these veterinary-specific drugs, and therefore, these compounds were excluded from our analysis.

With respect to classification of drug solubility, one of the challenges was the absence of the ideal canine gastric volume that can be used for estimating a dose number (Do). The Do used in the calculations for determining BCS criteria is determined by the formula:

where M is the dose strength of the tablet/capsule, V is the volume administered (defined as 250 mL for humans), and C is the drug’s solubility (mg/mL).

A volume of 250 mL, the typical volume of water consumed during human bioavailability studies, is too large to be appropriate to estimate the volume of water flush administered to a fasted dog. Therefore, other volumes were explored for the calculation of a Do. In one analysis, we used a volume of 6 mL because this is often the volume administered to dogs with an oral medication (a single “flush” with a 6-mL syringe). A volume of 6 mL also has been suggested as the residual volume in the empty canine stomach (1, , 25). Additional analysis was performed using 35 mL because it is equivalent to the 250 mL (~ one cup) when scaled to the size of an average Beagle dog (the breed used most often in oral drug absorption studies).

The relationships between canine estimates of F versus human BCS values, solubility, LogP, Do, and F values for humans were compared by linear regression. This was accomplished using the Proc Reg procedure in SAS (version 9.3). Both slope and intercepts were included in the regression equation. Confidence and prediction intervals about the regression line were set at alpha = 0.10 (90% intervals, 5% in each tail). The regression of Do on F was expressed relative to the natural logarithm of Do (LD). This evaluation was conducted both at Do values estimated at a volume of 6 mL (LD6) and 35 mL (LD35).

RESULTS

Oral absorption data were obtained for 50 drugs for which human and canine data were available (–; Papich, 1986, Pharmacokinetics of ranitidine and cimetidine in dogs, unpublished data; –; Papich, 1988, Pharmacokinetics of doxycycline in dogs after intravenous administration and oral administration of doxycycline hyclate and doxycycline monohydrate, unpublished data; –; Papich, 1986, Pharmacokinetics of lorazepam in dogs, unpublished data; –). Four drugs had conflicting data and therefore were listed twice to include both sets of data. For two drugs (, ), publications addressing BCS biowaivers for human formulations had data that conflicted with an official web site (27) or other published data (–). These drugs were listed twice to accommodate both data. Many of these drugs were administered to dogs as the human formulation or as a compounded product when there was no approved veterinary counterpart. For two drugs (furosemide and phenobarbital), two sets of canine values were considered because of duplication of published data. There was a relatively even distribution of drugs among the four BCS classes. There were 16 drugs from class I, 9 drugs from class II, 15 drugs from class III, and 10 drugs from class IV. As a percentage of drugs, this was a higher representation of class III and class IV drugs than the analysis of Takagi et al. ().

Table I summarizes the information used in our examination of the relationships between drug physicochemical parameters and the oral absorption performance in humans and dogs. For consistency, we relied primarily upon two sources of information for the BCS classification: Kasim et al. (), using their LogP-based estimates for our predictions of drug permeability, and Wu and Benet (). For those several compounds that were not contained within those two references, other published values were used (, , , ). Despite recognized diversity in some of the values across these various information sources, we concluded that since the magnitude of differences was small relative to the overall strength of the trends that were observed, any error that may have been introduced by the selection of publications would not influence the conclusions derived from our comparison.

Table I

Analysis of 50 Drugs in Humans and Dogs Using Solubility, Lipophilicity, and Comparison of Bioavailability (F), According to BCS Class (Four Drugs Are Listed Twice Because of Two Sets of Conflicting Data)

Drug nameMax dose (mg) humanIntrinsic solubility (mg/mL)DoLogPF humanBCS classDose dogs (mg)F (dog)Do (35 mL) dogsDo (6 mL) dogsReference
Acetaminophen5000.1200.890.8741400.6340.000233.333()
Amlodipine1010.040.290.743250.630.724.16()
Amoxicillin50040.5−0.580.9332720.7681.9411.33()
Atenolol10026.50.0150.160.5434000.830.432.52()
Azithromycin6000.012404.020.3422400.97685.714000()
Bisoprolol1010000.000041.940.81110.9140.000310.002()
Carvedilol250.01103.140.252110.14331.43183.33()
Cephalexin50012−0.670.9542400.576.8640.000()
Chlorpheniramine41000.000023.770.4111000.360.0290.17Papich, 1986, Pharmacokinetics of ranitidine and cimetidine in dogs, unpublished data,()
Cimetidine80060.530.790.62396.250.950.462.67()
Cimetidine80060.530.790.6231000.750.482.78(, )
Ciprofloxacin750100.31.320.642500.5840.714.17()
Clindamycin3001000.0122.160.8711500.72550.0430.250()
Clomipramine251000.0015.650.48156.20.160.0160.094()
Codeine601000.00241.450.53450.040.0130.075()
Cyclosporine1000.01402.950.282500.35142.86833.33(43)
Diazepam1010.042.9811400.1461.14296.67()
Diltiazem1201000.00482.640.381300.260.010.050()
Digoxin0.250.010.11.950.710.360.581.0296.000()
Dolasetron1001000.0040.0620.0681650.070.0190.11()
Doxycycline1000.14−3.660.9341000.36528.57166.67Papich, 1988, Pharmacokinetics of doxycycline in dogs after intravenous administration and oral administration of doxycycline hyclate and doxycycline monohydrate, unpublished data
Doxycycline230.8330.028−3.660.9311000.3650.080.49()
Fluoxetine50100.024.270.951200.720.0570.33(49)
Furosemide800.01320.740.6144000.771142.866666.67()
Furosemide800.01320.740.614400.38353.33666.67()
Gabapentin8001000.032−1.120.638500.80.241.42()
Hydralazine50400.0050.730.3531040.770.0740.43()
Hydroxyzine1000.01403.320.6242.50.72121.43708.33()
Ibuprofen8000.013203.140.8287.50.77250.001458.33()
Ketoprofen750.0515.93.310.921000.956.022326.80()
Levetiracetam75010400.00292.791150010.0140.080()
Linezolid60012.40.58142500.9667.1441.67(, )
Lorazepam20.080.12.390.9329.40.5983.3619.58Papich, 1986, Pharmacokinetics of lorazepam in dogs, unpublished data
Meloxicam150.0160.970.9722.5517.2942.50()
Metoclopramide100.0141.480.823230.47865.71383.33()
Metronidazole500100.2−0.460.991435.60.81.257.26()
Minocycline (HCl)100500.010.050.9531000.5030.080.5()
Morphine1062.50.0011.190.243150.0530.0070.040()
Naproxen500330.062.860.9911000.840.0870.51()
Phenobarbital1000.141.5211133.650.8838.19222.75()
Phenobarbital1000.141.5211364.50.97104.14607.50()
Phenytoin1000.01402.140.921550.36442.862583.33()
Piroxicam200.0233.483.060.924.215.2230.44()
Procainamide50050.40.880.8332500.851.438.33()
Propranolol90330.012.6511100.270.0090.051()
Ranitidine3001000.0120.630.27338.50.810.0110.064Papich, 1986, Pharmacokinetics of ranitidine and cimetidine in dogs, unpublished data
Rofecoxib500.01201.830.932500.26142.86833.33()
Sildenafil10010.42.110.381150.540.432.50()
Theophylline3008.330.144−1.030.9911600.910.553.20(, )
Tramadol50330.0062.520.73187.80.650.0760.44()

Do dose number (defined in the text), LogP log of octanol/water partition coefficient, F systemic bioavailability, BCS class Biopharmaceutical Classification System class as defined in the references cited in the text

When assessing the relationship between estimates of F in dogs and humans (Fig. 1), we observed that the slope significantly differed from zero (P = 0.0055). However, the corresponding low coefficient of determination (R2 = 0.15) indicates that human estimates of F very poorly predicted canine F values. Without considering drug physicochemical characteristics, the potential contribution of drug solubility and/or permeability on the observed interspecies inconsistencies could be determined (and hence, the reason behind our further examination of these data). The intercept of the regression line was 0.31, which might be construed as suggesting a trend toward higher canine drug bioavailability when the human oral drug bioavailability is very low. Indeed, of the nine drugs with human estimates of F less than 0.40, six were associated with canine F values equal to or greater than that observed in humans. However, given the very low R2 value, such conclusions should be construed as an overinterpretation of the observed canine/human relationship. Along with this caution is the observation of wider confidence and predictions toward the intercept.

The relationship between oral bioavailability (F) observed in humans versus dogs. Although there was much variability in the relationship between estimated values of F in dogs and people, a statistically significant correlation was observed (p = 0.0055). The confidence interval (as defined by the shaded region around the regression line) reflects variability both in slope and intercept attributable to the uncertainty in the estimated regression line. While the wider prediction interval likewise reflects the uncertainty of the data, it predicts the interval within which the regression will be contained (90% of the time) if the analysis were repeated using a comparable human and canine populations (i.e., relating the prediction of dog F from human F). The intercept (dog F observed when the human F = 0) is 0.31, indicating an overall trend toward a higher bioavailability in dogs at very small human values of F. The slope of the regression of dog versus human is 0.42

Despite the low overall correlation between human and canine F values, when segregated according to its BCS class relationship, patterns begin to emerge (Fig. 2a–d). We observe that as compared to that observed in BCS classes III and IV, many of the compounds contained in BCS classes I and II demonstrated a good correlation between F in dogs and humans. In terms of the percent of the listed compounds where the human and canine F values differed by no more than +/−20%, these were found to be 79, 64, 36, and 20% for BCS classes I–IV, respectively (Table II). For BCS class I compounds, we observed a trend toward a lower bioavailability in dogs as compared to that in humans (Fig. 2a). Similarly, the class IV compounds tended to exhibit a comparatively lower oral bioavailability in dogs (Fig. 2d). For the nonsteroidal anti-inflammatory drugs (NSAIDs), all but one showed similar F values in dogs and humans (four were in class II or IV), which is consistent with their BCS classification of a low solubility compound being primarily a function of their behavior in acid. A surprisingly high number of drugs in class II (Fig. 2c) showed similar or higher F values in dogs compared to humans. Several of these compounds were weak bases.

Comparison of canine/human absolute bioavailability (F) as a function of human BCS classification: a BCS class I compounds, b BCS class II compounds, c BCS class III compounds, and d BCS class IV compounds. The hatched line represents the theoretical line of unity

Table II

Comparison of Drugs According to Bioavailability in Humans, Dogs, Therapeutic Class, pKa, and Metabolism

Comparative F, dog versus humanaBCSDrugHuman F (dose mg)Dog F (dose mg)Therapeutic classpKaFirst-pass lossCyp enzymeReference
Similar1Bisoprolol0.8 (10)0.91 (11)Beta-1 adrenergic blocker9.5 (base)NoCyp3A4, some Cyp2D6DrugBank
Similar1Chlorpheniramine0.41 (4)0.36 (100)Alkylamine antihistamine9.2 (base)YesCYP2D6http://cadd.suda.edu.cn/admet/compound/detail/573; ()
Similar1Clindamycin0.87 (300)0.73 (150)Lincosamide7.7 (base)NoCYP3A4http://cadd.suda.edu.cn/admet/compound/detail/1350; Wynalda et al. ()
Similar1Digoxin0.7 (0.25)0.58 (0.36)Cardiotonic glycosideNoPgp may limit absorption. Minimal metabolismhttp://www.rxlist.com/lanoxin-tablets-drug/clinical-pharmacology.htm
Similar1Diltiazem0.38 (120)0.26 (30)Ca channel blocker7.7 (base)Yes (Cyp3A4)Cyp3A4DrugBank, druginfosys.com
Similar1Dolasetron0.068 (100)0.07 (65)Serotonin 5-HT3 receptor antagonistNAYesCYP2D6, CYP3A4(43); http://cadd.suda.edu.cn/admet/compound/detail/878
Similar1Levetiracetam1.0 (750)1.0 (500)Anticonvulsant (binds to synaptic vesicle)NANoEnzymatic hydrolysisDrugBank, druginfosys.com
Similar1Linezolid1.0 (600)0.97 (250)Anti-infective1.7 (base)NoPrimarily renal elimination of unchanged drug(, ); http://www.fda.gov/ohrms/dockets/ac/00/backgrd/3597b1bb.pdf
Similar1Metronidazole0.99 (500)0.80 (435.6)Antiprotozoal, antibacterial (anaerobe)2.6 (base)NoPrimarily CYP2A6, some CYP3A4, 5, and 7DrugBank, PharmacoKinetics Knowledge Base; ()
Similar1Naproxen0.99 (500)0.84 (100)NSAID4.15 (acid)NoCYP2c8, 2c9, 1A2, UDP-glucuronosyltransferaseDrugBank
Similar1Phenobarbital1.0 (100)0.88–0.97 (133.65 to 364.5)Nonselective central nervous system depressant7.3 (acid)NoMostly CYP2C19DrugBank, PharmacoKinetics Knowledge Base
Similar1Sildenafil0.38 (100)0.54 (15)Phosphodiesterase type 5 inhibitor8.2 to 9.6 (base)YesCYP3A4, also CYP2C9DrugBank, http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-Public_assessment_report/human/001080/WC500068025.pdf
Similar1Theophylline0.99 (300)0.91 (160)Phosphodiesterase inhibitor8.6 (base)NoCYP1A2DrugBank, PharmacoKinetics Knowledge Base
Similar1Tramadol0.73 (50)0.65 (87.8)Opioid9.41 (base)NoCYP3A4 and CYP2D6DrugBank, druginfosys.com
Not Similar1Clomipramine0.48 (25)0.16 (56.2)Antidepressant9.38YesCYP2C19, CYP3A4, and CYP1A2DrugBank, PharmacoKinetics Knowledge Base, http://www.pharmgkb.org/pathway/PA165960076
Not Similar1Diazepam1.0 (10)0.15 (40)Benzodiazepine3.3No (human)CYP2C19 and CYP3A4DrugBank, PharmacoKinetics Knowledge Base
Not similar1Doxycycline hylate0.93 (100)0.37 (100)Tetracycline3.5, 7.7, 9.5No (human)Negligiblehttp://cadd.suda.edu.cn/admet/compound/detail/1399
NOT SIMILAR1Fluoxetine0.95 (50)0.72 (20)Serotonin uptake inhibitor8.7 (base)YesCYP2D6DrugBank, PharmacoKinetics Knowledge Base
NOT SIMILAR1Propranolol1.0 (90)0.27 (10)Beta blocker9.5Yes (saturable)CYP2D6, 1A2, and UGTDrugBank, PharmacoKinetics Knowledge Base
Similar2Carvedilol0.25 (25)0.14 (11)Beta blocker7.6 (base)YesCYP2D6, CYP2C9DrugBank, PharmacoKinetics Knowledge Base
Similar2Cyclosporine0.28 (100)0.35 (50)Immune-suppressant6.9 (base)YesCyp3A4(, )
Similar2Hydroxyzine0.60 (100)0.72 (42.5)Antihistamine1.8, 2.1, 7.1NoCyp3A4/5Ucerax (hydroxyzine hydrochloride) 25 mg film-coated tablets. Summary of product characteristics. Irish Medicines Board. Retrieved 9 February 2014, http://web.squ.edu.om/med-Lib/MED_CD/E_CDs/A%20Practical%20Guide%20to%20Contemporary%20Pharmacy%20Practice/pdf/pKa-table.pdf
Similar2Ibuprofen0.80 (800)0.77 (87.5)NSAID4.9 (acid)NoCYP2C9http://cadd.suda.edu.cn/admet/compound/detail/277
Similar2Ketoprofen0.9 (75)0.9 (100)NSAID4.45 (acid)NoGlucuronidationDrugBank
Similar4Meloxicam0.97 (15)1.0 (2.55)NSAID4.47 (acid)NoCYP2C9DrugBank, PharmacoKinetics Knowledge Base
Similar2Piroxicam0.9 (20)1.0 (4.2)NSAID6.3 (acid)NoCYP2C9DrugBank
Similar3Amlodipine0.74 (10)0.63 (25)Calcium channel blocker8.6 (base)NoCYP3A4DrugBank, PharmacoKinetics Knowledge Base, http://www.pfizer.ca/en/our_products/products/monograph/212
Similar3Amoxicillin0.93 (500)0.83 (400)Beta lactam2.4, 7.4, 9.6NoNegligiblehttp://cadd.suda.edu.cn/admet/compound/detail/1089
Similar3Cimetidine0.76 (800)0.95 nH2-receptor antagonist6.8 (base)NoCimetidine/H+ exchange mechanismPiyapolrungroj et al. ()
Similar3Procainamide0.83 (500)0.85 (250)Sodium channel blocker9.4 (base)NoCYP2D6, CYP3A4DrugBank, PharmacoKinetics Knowledge Base
Similar4Ciprofloxacin0.60 (750)0.58 (250)Antimicrobial6.09 (acid) and 8.89 (base)NoSeveral (including CYP1A2)DrugBank, PharmacoKinetics Knowledge Base

BCS class Biopharmaceutical Classification System class as defined in the references cited in the text, F systemic bioavailability, Cyp enzyme cytochrome P450 enzyme, NSAID nonsteroidal anti-inflammatory drug

aDefinition of similar versus not similar: “Similar” is as any human/canine value of F that differed by no more than +/−20%. The 20% value was based upon the traditional bioequivalence (BE) criteria (linear scale) of equivalence being +/−20%. “Not similar” was defined as outside this range

As compared to that seen for BCS classes I and II, far fewer compounds exhibited comparable human versus dog bioavailability for BCS classes III and IV. With regard to the latter two classes, there was no obvious pattern identified such that very high or very low oral absorption could be correlated with values of F when comparing dogs to humans. For example, for class III drugs (Fig. 2c), there was somewhat of an even distribution above and below a line of unity. There were examples of some compounds that were more bioavailable in humans than in dogs (e.g., codeine), while others were more bioavailable in dogs than humans (e.g., ranitidine).

One of the potential sources of interspecies bias is the difference in gastric volume versus dose. For this reason, we estimated the Do, using a volume of 6 or 35 mL (representing a range of volumes administered in studies where dogs are administered a water flush after oral dose administration). As shown in Fig. 3a (6 mL volume) and b (35 mL volume), no obvious association between the calculated Do (expressed as LD) and F could be identified. This is evidenced both by low R2 values (0.031 and 0.029 and for 35 mL (LD35) and 6 mL (LD6), respectively) and the lack of statistical significance when evaluating the slope of the regression line (P = 0.23 and 0.22 for 3a and b, respectively), indicating that these slopes do not significantly differ from zero. Furthermore, the similarity in width of the confidence and prediction intervals at the upper and lower portions of the profile suggests that the error about the regression line is similar across the range of LD values obtained in this study.

The influence of the log-transformed dose number (LD) on the estimated value of F in dogs. a The relationship defined by volume of 6 mL (LD6). b The relationship defined by a volume of 35 mL (LD35). Interpretation of confidence versus prediction intervals corresponds with that previously described for Fig. 1

Figure 3a, b illustrates that regardless of whether a volume of 6 or 35 mL was used in the analysis, there was little influence of fluid volume on Do above, or below, 1.0. This indicates that either volume could adequately reflect oral dose solubility as a function of administered dose for most drugs administered to dogs. Using a Do of 1.0 as the cutoff between low and high solubility (as used for the human BCS), only seven drugs (gabapentin, cimetidine, amlodipine, ciprofloxacin, sildenafil, theophylline, and atenolol) would be classified as high solubility (Do < 1) using a volume of 35 mL, but low solubility (Do > 1) using a volume of 6 mL.

The influence of the lipophilicity (expressed as LogP) on F was examined in Fig. 4. When considering all of the drugs included in this analysis, no association could be identified between LogP and F (R2 = 0.0027). The large P value for the slope of the regression line (P = 0.72) indicates that the slope defining the regression of LogP on canine F is not significantly different from zero. Furthermore, similarities in the width of the confidence and prediction intervals at the upper and lower portions of the profile suggest that that the error about the regression line was similar across the range of LogP values. However, limiting our assessments to those drugs with F > 0.80 (Fig. 5), most had LogP values within the range of 0–4, suggesting transcellular absorption. The only compounds showing high F values but LogP < 0 in dogs were gabapentin, amoxicillin, metronidazole, and theophylline. The Do values in these four drugs ranged from below 0.001 to over 1000.

LogP value versus F value in dogs for 50 drugs. LogP is the experimentally determined lipid partition coefficient; the F value is the bioavailability in dogs. The estimated canine F value when LogP is zero is 0.62. The corresponding slope of the line is −0.008. The large P value indicates that this is not a significant correlation, and therefore, the negative slope should not be construed as being indicative of any true relationship. Interpretation of confidence versus prediction intervals corresponds with that previously described for Fig. 1

High bioavailability (approximately 80%) drugs in dogs compared to dose number and LogP. (Dose number and LogP were previously defined.) Four drugs (shown with open diamonds on the left side of the figure) were exceptions because they had LogP < 0, but good bioavailability: metronidazole, which is a small molecule absorbed paracellularly in dogs; gabapentin and amoxicillin, which are probably absorbed via intestinal transporters; and theophylline, which can be absorbed from the large intestine (colon)

To identify some commonality between agents exhibiting high human/canine correlations, we examined the pKa, mechanism of drug elimination (human), and whether or not a human first-pass drug loss was reported (Table II). The vast majority of compounds with similar values of F in dogs and humans were also those compounds with negligible first-pass drug loss. Furthermore, in the three out of four BCS class I compounds where human and canine F values were discordant, those compounds were reported to have a high human first-pass drug loss.

DISCUSSION

Whether we are providing values for F, solubility, permeability, or BCS classification, all values need to be viewed from the perspective as estimates derived under a specific set of experimental conditions. Thus, there are occasions where, for example, a compound was estimated as being within one BCS classification in one reference but as a different class for another reference (e.g., ciprofloxacin). The tables available from Takagi et al. () show some of these discrepancies. Similarly, values of F reported within this manuscript reflect a distinct set of experimental conditions, and therefore, there can be differences in reported estimates elsewhere, depending upon study conditions and population. For example, we can consider the compound amlodipine. In one study, the human oral bioavailability was stated to be 63% (). However, in the Pfizer monograph for amlodipine besylate (http://www.pfizer.ca/en/our_products/products/monograph/212), the absolute bioavailability of an oral administration ranged between 64 and 90%. Accordingly, on an individual compound basis, the estimated ratio of dog and human F values provided in this manuscript should be considered a single point within a distribution of potential estimates. That said, when viewed across a diverse library of compounds (as we have done in this manuscript), these values provide a tool for examining variables that can be used to examine interspecies relationships in drug absorption.

Highly soluble and highly permeable compounds will likely exhibit similar oral drug bioavailabilities unless the drug exhibits differing extent of first-pass metabolism in dogs versus humans. Nevertheless, we cannot conclude that a drug’s BCS classification will necessarily translate across species. BCS solubility classifications are traditionally based upon a calculation of the Do, which assumes a dissolution volume of 250 mL (approximately one cup of water). That volume is far greater than the gastric volume of the fasted dog. Typically, studies performed in dogs will include a water flush of 6 or 12 mL because, for practical reasons, these are common plastic syringe sizes. We included a high value of 35 mL in our analysis because this represents the equivalent to the 250 mL (~ one cup) in humans scaled to the size of an average Beagle dog (the breed used most often in oral drug absorption studies). Accordingly, compounds considered highly soluble in a human population may not meet the criteria for highly soluble in dogs. Regardless of whether we consider the canine gastric volume to be 6 or 35 mL, we found that the solubility classification would be affected for only a few compounds (Fig. 3a, b). Using a Do cutoff of 1.0 for distinguishing between low or high solubility compounds, only seven drugs, gabapentin, cimetidine, amlodipine, ciprofloxacin, sildenafil, theophylline, and atenolol, would have had a different solubility classification as a function of different volumes used for these calculations. In some cases, this solubility difference may impact oral drug absorption. For example, in the case of ciprofloxacin, Papich () observed that differences in the volume administered with the oral dose may indeed affect F in dogs. When ciprofloxacin tablets were administered with a volume that decreased the Do below 1, oral absorption was better than when the Do was >1. This observation points to the importance of dosing regimens and study design when interpreting study data.

Another challenge facing interspecies BCS extrapolations is that by convention, a drug is classified as highly soluble based upon the highest administered dose. In the case of diazepam, the drug itself is poorly soluble, but may be listed as a human BCS class I compound because of the low administered dose. The importance of considering dose was underscored with diazepam where our cited canine investigations used a dose of 40 mg (i.e., approximately 4 mg/kg). This is in contrast with a 10-mg dose in humans (i.e., approximately 0.14 mg/kg for a 70-kg person). Thus, although included in the list of BCS class I compound for this analysis, it would have been more appropriate to have classified diazepam as a BCS class II drug (at least for the sake of the canine investigation). Diazepam was in the lower right quadrant of the group of class I drugs (Fig. 2a), and considering the low solubility of diazepam, it is not surprising that its oral bioavailability was very poor in dogs where tablet dissolution is further compromised by a rapid GI transit time (vide infra).

The BCS classification is influenced by the ability of the highest dose strength to be fully solubilized in 250 mL (~ one cup) of aqueous media over the pH range of 1–7.5. However, for low solubility compounds, interspecies differences in absorbable dose may also be impacted by the composition of the GI fluids. In this regard, the unique composition of canine GI fluid can lead to differences in canine-human in vivo tablet dissolution ().

Because dissolution impacts oral bioavailability, it is important to distinguish between criteria used to evaluate the inherent solubility of the active pharmaceutical ingredient (API) versus that used to characterize in vivo dissolution of a dosage form. As compared to an assessment of the API, the dissolution rate (DR) of any formulated product is a function of its available surface area (A), the diffusion coefficient (D) of the drug (i.e., its ability to move from the undissolved portion of the API to the surrounding dissolution medium, the effective boundary layer thickness (h, which is the water that physically surrounds the undissolved API), the saturation concentration of the API under the conditions of the dissolution test (Cs), the amount of drug already dissolved (Xd), and the volume of fluid (V) within which the dissolution must occur (). From an interspecies perspective, differences in GI fluid volume and composition can have an effect on most of these variables. These interrelationships are described by the Noyes-Whitney equation ():

These considerations will be particularly important for low solubility compounds and may further complicate efforts to generate interspecies comparisons based upon the bioavailability of solid oral dosage forms.

Another challenge facing an examination of canine BCS criteria is that of defining what constitutes a high versus low permeability compound. Because there are no commercially available canine cell lines (), in the absence of in vitro permeability data, we have relied upon LogP values as the basis for estimating a canine BCS permeability cutoff value. Our selection of LogP values was founded upon published reports where LogP was used as a surrogate for human permeability studies (–). These referenced studies used metoprolol, with a LogP = 1.72 as the reference standard for the cutoff between high/low permeability. Drugs with LogP > 1.72 are classified as highly permeable, while those with LogP < 1.72 are classified as low permeability (, ). Our literature search did not produce any canine pharmacokinetic data for oral metoprolol. However, we did have information on a similar β-adrenergic antagonist, bisoprolol, which has chemical characteristics similar to that of metoprolol (BCS class 1, low Do, and LogP 1.94) and greater than 90% oral absorption in dogs (), suggesting that we could rely upon the human LogP cutoff of 1.72 for our canine assessments.

We failed to observe a correlation between a LogP cutoff value of 1.72 versus those drugs having high or low F values in dogs (Fig. 4). In fact, a line drawn at a LogP of 1.72 would practically split this group of drugs down the middle. That is, many drugs were found to be highly bioavailable (and therefore presumably highly permeable) whether or not they had LogP values above or below 1.72. Likewise, we identified several BCS class III drugs (high solubility, low permeability) with F values as high, or higher in dogs as compared to humans (Fig. 2c).

In the absence of first-pass drug loss, further study will help to determine whether or not the use of LogD (which is based upon both ionized and unionized species, i.e., is a pH-dependent value) rather than LogP would have been a more appropriate metric because LogD values better reflect the impact of GI pH on human-canine permeability differences. For compounds with molecular weights ranging from 165 to 644, the relationship between the Caco-2 cell-based apparent permeability (Papp) and LogD values were described by a bell-shaped relationship (). Highly permeable compounds (i.e., those with Papp values of 100–340 nm s−1) had LogD values ranging between 0 and 3, while low permeability compounds had values of either less than −1.5 or greater than 4.5. However, Kramer () observed that the reliability of LogD for estimating permeability appears to diminish when the molecular weight exceeds 500, and additional variables impacting permeability (e.g., hydrogen bonding properties, molecular size and shape, polarity, and flexibility) may need to be examined.

When considering the compounds where we have identified dog-human disparities in F, presystemic drug metabolism is likely to be an important factor to consider. This was seen in the drugs that fell below the line of unity in Fig. 2a–d, where several of these agents are known to be highly metabolized in dogs. There are also compounds (cyclosporine, carvedilol, dolasetron, and diltiazem) that were associated with high first-pass drug loss but had similar human-canine values of F. Therefore, an identification of pathways (e.g., CYP450 metabolizing enzymes, uptake and efflux transporters) responsible for the presystemic drug loss may help predict those drugs for which there is likely to be interspecies bioavailability differences.

Presystemic drug loss is a likely reason for some of the differences in the interspecies relationship described by Chiou et al. () as compared to that reported in this current study. In particular, canine oral bioavailability in this study was either similar to or lower than that reported by Chiou and colleagues. They () reported values based upon urinary concentrations of total drug, but the studies used in our analysis analyzed only the parent compound, typically using HPLC methods. We believe that the trend toward lower bioavailability in our dataset likely reflects differences in gut or hepatic presystemic metabolism in the dog.

Although one ordinarily anticipates a low F for poorly permeable drugs, there were two BCS class III and IV compounds with human F > 0.90 (amoxicillin and cephalexin). Despite their poor membrane permeability, both compounds undergo active transport via peptide transporter 1 (PEPT1) (–). Thus, enterocyte penetration via facilitated/active rather than passive processes will result in an extent of absorption that cannot be predicted solely on the basis of BCS classification criteria. Moreover, numerous BCS class III and IV compounds were associated with an F > 0.80 in dogs but not in humans. It is undetermined why these drugs defined as poorly permeable exhibited such high F values in dogs. Because the dog intestine is anatomically shorter than that of humans, in order to absorb nutrients, the canine intestine may have evolved a greater capacity for intestinal uptake transport as compared to that of humans. Less active intestinal efflux transport also is possible. For low molecular weight compounds, there is also greater paracellular absorption for some compounds in dogs than humans () (vide infra).

For furosemide, there was higher bioavailability from a 400-mg dose (F 0.77) compared to a tenfold lower dose of 40 mg (F 0.38). Class III and IV drugs ordinarily typically do not exhibit saturable drug efflux (). But for this class IV compound, efflux transport apparently can be saturated at high doses, thus increasing its oral bioavailability (–). Therefore, it would seem likely that at the high doses of furosemide administered in the canine study, an increase in enterocyte concentrations produced a saturation of efflux transporters and a corresponding larger F.

When drugs with relatively high F values in dogs were considered (Fig. 5), most of those compounds were associated with LogP values >0. The only drugs in this study that showed both high F values in dogs and LogP < 0 were gabapentin, amoxicillin, metronidazole, and theophylline—all class III drugs that were in the upper right quadrant of Fig. 2c. In these cases, the high F in dogs, despite low LogP, can be explained by other factors. Gabapentin and amoxicillin rely on intestinal influx transporters for their absorption. Metronidazole and theophylline are small molecules (molecular weight 171 and 198, respectively) that can be absorbed via paracellular pathways. Drugs of low molecular weight are likely to be absorbed from the intestine via the paracellular pathway rather than by transcellular diffusion. Dogs have larger pore size and a greater frequency of pores in the intestine compared to that observed in other species (). This difference in pore number and diameter may have contributed to the observed greater oral absorption irrespective of the compound’s Peff value. Furosemide is another example of a drug for which paracellular absorption has a significant impact (, ) and where the differences between F in humans and dogs may reflect the competing effects of paracellular versus transcellular absorption and efflux transport of those molecules that did successfully enter into the enterocyte.

Some of the BCS class III compounds such as morphine and codeine (high solubility, low permeability) had lower F in dogs than in humans (Fig. 2c). For these compounds, the marked interspecies difference in oral bioavailability is the result of a high first-pass metabolism in dogs (, ), a characteristic of oral opiates in dogs that is not as prominent in humans. Other oral opiates not included in this analysis also have essentially zero oral systemic availability in dogs (, ). For these drugs, systemic clearance exceeds liver blood flow and there may be extrahepatic metabolism contributing to the high first-pass metabolism.

For lipophilic compounds (BCS class II and IV), solubilization can be significantly impacted by bile salt composition and excretion. If the bile salt-drug interactions result in micelle formation, the resulting encapsulation can lead to a decrease in drug transport, limiting the ability of free drug to move across a biological membrane (). The formation of micelles can either increase or decrease drug absorption, depending upon the magnitude of its impact on solubility versus permeability (). A micellar-induced increase in the solubility of lipophilic compounds (due to endogenous, food-associated, or formulation-associated surfactants) can increase or decrease drug intestinal permeability () by reducing the partition coefficient between the intraluminal fluid and the biological membrane (). Thus, the pH and composition of GI fluids can be pivotal to dog-human differences in the absorption of lipophilic molecules.

The analysis presented in this paper reveals some challenges and pitfalls of relying on the human BCS parameters to predict performance of oral drug products in dogs. Because the BDDCS also takes into consideration presystemic drug metabolism, the combination of the BCS and BDDCS criteria for evaluating oral drugs in dogs may be a better approach to examine human/canine differences in oral bioavailability than the use of a single classification system. This approach is consistent with the perspective explained by Shugarts and Benet () where they note that the BDDCS considers the major route of elimination as an important factor in classification instead of permeability. As emphasized by Shugarts and Benet (), “BCS cannot predict absorption,” which is what we observed in this study. The BDDCS divides drugs into four classes based on solubility (high or low, as in the BCS), and whether or not the drug has low, or extensive metabolism. Because permeability data is difficult to acquire for medications in dogs, and because the LogP reference values used to assess human GI permeability do not appear to predict oral absorption for dogs, a system such as the BDDCS or some new system for dogs should be explored.

We are continuing in our effort to examine the specific factors that explain the observed human-canine differences in oral drug absorption. When considering sources of potential interspecies divergence, the important physiologic variables include GI fluid pH and composition, intestinal transit time, mucosal surface area, and the size and density of intestinal intercellular pores (–). These factors are particularly influential in the absorption of low solubility and/or low permeability drugs and may be helpful to predict interspecies differences in food effects (). Further considerations into metabolic pathway and influx/efflux transporter activity needs to be incorporated into our overall assessment. It is with these points in mind that we have summarized the important variables to incorporate into our human-canine interspecies extrapolations (Table II).

CONCLUSION

This investigation showed that applying the same BCS criteria to dogs and humans can be problematic. At least in part, when attempting to designate a BCS classification for dogs, there is a need to develop canine-specific solubility and permeability assessments. Ultimately, even when the necessary in vitro methods for estimating canine drug solubility and permeability have been developed, canine-human physiological differences can result in marked differences in systemic absorption due to transporter functions, drug metabolism, and the leakier canine intestinal membranes.

Clearly, there remains much work to be done in order to improve our ability to predict drug absorption in the dog when based upon preliminary drug physicochemical characterization and an interspecies extrapolation of in vivo PK information. Efforts to predict drug absorption (or to understand the causative factors impacting interspecies differences in F) is dependent upon a wide array of variables including API solubility and permeability (BCS), formulation factors, and physiological variables (including regional permeability differences, which could differ between species), GI pH, luminal and mucosal enzymology, and intestinal motility, first-pass drug metabolism, and transporter activity (–). It is for this reason that a blending of BCS and the BDDCS may provide far better predictions of canine versus human drug absorption characteristics than would either classification system alone. Comparing drug absorption characteristics in dogs and humans, while important for interspecies extrapolations and for formulation development, also provides valuable insights into the variables that can influence drug absorption (and interindividual differences) that can occur in the presence of human or canine GI pathologies, breed potential breed effects on canine drug absorption, or the changes in drug absorption that may occur in the geriatric dog or human population. Such predictions are predicated largely upon the development of mechanistic (in silico) models. By understanding the impact of these critical variables on the rate and extent of drug absorption, these models can be positioned for use in predicting the canine to human (and vice versa) differences in oral drug absorption and in vivo product performance.

Our ongoing efforts are based upon the use of this drug list to explore published human-canine differences in drug metabolism, along with an evaluation of other potential variables. We anticipate that through these efforts, we will obtain a better appreciation of the pivotal factors dictating in vivo product absorption and determine potential sources of error when attempting to extrapolate information obtained in humans to support in silico predictions of in vivo drug absorption characteristics in dogs.

At the time this manuscript was prepared, we did not have access to the paper published by Musther et al. (). Their paper included 125 paired datasets comparing F between dogs and humans and included many of the same studies that were used in our analysis. As we report here, they also found a poor correlation in F for drugs between dogs and humans with an R value of 0.37. Musther et al. () did not include BCS criteria in their analysis, but instead, separated compounds into acidic, basic, neutral, or zwitterions. This improved the prediction slightly for acidic drugs, some of which were likely BCS class II compounds. A prediction model of human F from canine values, based on a linear regression model, resulted in wide prediction intervals with a low concordance correlation coefficient (precision of the prediction), confirming the lack of agreement between human and dog F values. As in our studies, they found that Beagle dogs were represented more often than other dog breeds for bioavailability determination in most studies (66%). We encountered similar problems as described by Musther et al. () when extracting data from published studies. Some studies provided full details of the methods and results and others provided limited data. They concluded, as we did, that metabolic differences between species could play a more important role in defining disparities between human and animal drug bioavailability.

ACKNOWLEDGMENTS

The authors would like to thank Robyn Martinez for her help collecting data for this manuscript. They also appreciate the support from the United States Pharmacopeia in the early stages of this project. The authors also acknowledge the advice and recommendations from Dr. G.L. Amidon and Dr. James Polli in the analysis of these data.

Conflicts of Interest

The authors have no conflicts of interest to disclose.

Apr 21, 2018 - Comedy A comedic spoof based on the worldwide phenomenon, The Twilight Saga. The Twilight Saga: Breaking Dawn – Part 1 (2011) full. Twilight breaking dawn part 1 full movie. Feb 26, 2018 - Movie Overview The Twilight Saga Breaking Dawn Part 1 2011 Dual Audio Movie Free Download Bella Swan is preparing for her wedding.

Contributor Information

Mark G. Papich, Email: ude.uscn@hcipapgm.

Marilyn N. Martinez, Email: vog.shh.adf@zenitram.nyliram.

REFERENCES

1. Martinez MN, Papich MG, Riviere JE. Veterinary application of in vitro dissolution data and the biopharmaceutics classification system. Stimuli to the revision process. Pharmacopeial Forum (United States Pharmacopeial Convention) 30(6): 2–10, 2004.
2. Amidon GL, Lennernäs H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995;12:413–20. doi: 10.1023/A:1016212804288. [PubMed] [CrossRef] [Google Scholar]
3. Lindenberg M, Kopp S, Dressman JB. Classification of orally administered drugs on the World Health Organization model list of essential medicines according to the biopharmaceutics classification system. Eur J Pharm Biopharm. 2004;58:265–78. doi: 10.1016/j.ejpb.2004.03.001. [PubMed] [CrossRef] [Google Scholar]
4. Lennernäs H, Abrahamsson B. The use of biopharmaceutic classification of drugs in drug discovery and development: current status and future extension. J Pharm Pharmacol. 2005;57:273–85. doi: 10.1211/0022357055263. [PubMed] [CrossRef] [Google Scholar]
5. Kasim NA, Whitehouse M, Ramachandran C, Bermejo M, Lennernas H, Hussain AS, et al. Molecular properties of WHO essential drugs and provisional biopharmaceutical classification. Mol Pharm. 2004;1:85–96. doi: 10.1021/mp034006h. [PubMed] [CrossRef] [Google Scholar]
6. Takagi T, Ramachandran C, Bermejo M, Yamashita S, Yu LX, Amidon GL. A provisional biopharmaceutical classification of the top 200 oral drug products in the United States, Great Britain, Spain, and Japan. Mol Pharma. 2006;3(6):631–43. doi: 10.1021/mp0600182. [PubMed] [CrossRef] [Google Scholar]
7. Wu CY, Benet LZ. Predicting drug disposition via application of BCS: transport/absorption/elimination interplay and development of a biopharmaceutics drug disposition classification system. Pharm Res. 2005;22:11–23. doi: 10.1007/s11095-004-9004-4. [PubMed] [CrossRef] [Google Scholar]
8. Benet LZ. The role of BCS (biopharmaceutics classification system) and BDDCS (biopharmaceutics drug disposition classification system) in drug development. J Pharm Sci. 2013;102(1):34–42. doi: 10.1002/jps.23359.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
9. FDA Guidance For Industry, CDER, “Waiver of in vivo bioavailability and bioequivalence studies for immediate-release solid oral dosage forms based on a biopharmaceutics classification system”, 2000.
10. Heikkinen AT, Fowler S, Gray L, Li J, Peng Y, Yadava P, et al. In vitro to in vivo extrapolation and physiologically based modeling of cytochrome P450 mediated metabolism in beagle dog gut wall and liver. Mol Pharm. 2013;10(4):1388–99. doi: 10.1021/mp300692k. [PubMed] [CrossRef] [Google Scholar]
11. Box KJ, Völgyi G, Baka E, Stuart M, Takács-Novák K, Comer JE. Equilibrium versus kinetic measurements of aqueous solubility, and the ability of compounds to supersaturate in solution—a validation study. J Pharm Sci. 2006;95(6):1298–307. doi: 10.1002/jps.20613. [PubMed] [CrossRef] [Google Scholar]
12. Venkatesh S, Li J, Xu Y, Vishnuvajjala R, Anderson BD. Intrinsic solubility estimation and pH-solubility behavior of cosalane (NSC 658586), an extremely hydrophobic diprotic acid. Pharm Res. 1996;13(10):1453–9. doi: 10.1023/A:1016059008464. [PubMed] [CrossRef] [Google Scholar]
13. Takács-Novák, K. Solubility-pH profile of drugs. Experiences and surprises in logS measurements, Semmelweis University, Budapest, Phys Chem FORUM 9 September 17, 2010.
14. Teksin ZS, Seo PR, Polli JE. Comparison of drug permeabilities and BCS classification: three lipid-component PAMPA system method versus Caco-2 monolayers. AAPS J. 2010;12(2):238–41. doi: 10.1208/s12248-010-9176-2.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
15. Buckley ST, Fischer SM, Fricker G, Brandl M. In vitro models to evaluate the permeability of poorly soluble drug entities: challenges and perspectives. Eur J Pharm Sci. 2012;45(3):235–50. doi: 10.1016/j.ejps.2011.12.007. [PubMed] [CrossRef] [Google Scholar]
16. Balimane PV, Chong S, Morrison RA. Current methodologies used for evaluation of intestinal permeability and absorption. J Pharmacol Toxicol Methods. 2000;44:301–12. doi: 10.1016/S1056-8719(00)00113-1. [PubMed] [CrossRef] [Google Scholar]
17. Pelkonen O, Boobis AR, Gundert-Remy U. In vitro prediction of gastrointestinal absorption and bioavailability: an experts’ meeting report. Eur J Clin Pharmacol. 2001;57:621–9. doi: 10.1007/s002280100369. [PubMed] [CrossRef] [Google Scholar]
18. He Y-L, Murby S, Warhurst G, Gifford L, Walker D, Ayrton J, et al. Species differences in size discrimination in the paracellular pathway reflected by oral bioavailability of poly(ethylene glycol) and D-peptides. J Pharm Sci. 1998;87(5):626–33. doi: 10.1021/js970120d. [PubMed] [CrossRef] [Google Scholar]
19. Rubas W, Jezyk N, Grass GM. Comparison of the permeability characteristics of a human colonic epithelial (Caco-2) cell line to colon of rabbit, monkey, and dog intestine and human drug absorption. Pharm Res. 1993;10:113–8. doi: 10.1023/A:1018937416447. [PubMed] [CrossRef] [Google Scholar]
20. Davis JL, Little D, Blikslager AT, Papich MG. Mucosal permeability of water-soluble drugs in the equine jejunum: a preliminary investigation. J Vet Pharmacol Ther. 2006;29(5):379–85. doi: 10.1111/j.1365-2885.2006.00757.x. [PubMed] [CrossRef] [Google Scholar]
21. Neirinckx E, Vervaet C, Michiels J, De Smet S, Van Den Broeck W, Remon JP, et al. Feasibility of the Ussing chamber technique for the determination of in vitro jejunal permeability of passively absorbed compounds in different animal species. J Vet Pharmacol Ther. 2011;34(3):290–7. doi: 10.1111/j.1365-2885.2010.01218.x. [PubMed] [CrossRef] [Google Scholar]
22. Kramer, S. Absorption prediction from physicochemical parameters. Pharmaceutical Science and Technology, 1999, 2, 373±380. [PubMed]
23. Martinez MN, Amidon GL. A mechanistic approach to understanding the factors affecting drug absorption: a review of fundamentals. J Clin Pharmacol. 2002;42(6):620–43. doi: 10.1177/00970002042006005. [PubMed] [CrossRef] [Google Scholar]
24. Martinez MN, Papich MG. Drug solubility classification in the dog. J Vet Pharmacol Ther. 2012;35(Suppl. 1):87–91. doi: 10.1111/j.1365-2885.2012.01373.x. [PubMed] [CrossRef] [Google Scholar]
25. Apley M, Crist B, Gonzalez MA, Hunter RP, Martinez MN, Modric S, et al. Solubility criteria for veterinary drugs. USP stimuli article. Rockville: United States Pharmacopeia Convention; 2012. [Google Scholar]
26. Chiou WL, Jeong HY, Chung SM, Wu TC. Evaluation of using dog as an animal model to study the fraction of oral dose absorbed for 43 drugs in humans. Pharm Res. 2000;17:135–40. doi: 10.1023/A:1007552927404. [PubMed] [CrossRef] [Google Scholar]
27. Therapeutic Systems Research Laboratories (TSRL), Inc. Biopharmaceutical Classification System (BCS). Ann Arbor, Michigan. http://www.tsrlinc.com/resources/services/. Accessed 24 Jul 2014.
28. Grabowski T, Jaroszewski JJ. Bioavailability of veterinary drugs in vivo and in silico. J Vet Pharmacol Ther. 2009;32(3):249–57. doi: 10.1111/j.1365-2885.2008.01034.x. [PubMed] [CrossRef] [Google Scholar]
29. Sagara K, Mizuta H, Ohshiko M, Shibata M, Haga K. Relationship between the phasic period of interdigestive migrating contraction and the systemic bioavailability of acetaminophen in dogs. Pharm Res. 1995;12(4):594–8. doi: 10.1023/A:1016270417009. [PubMed] [CrossRef] [Google Scholar]
30. Stopher DA, Beresford AP, Macrae PV, Humphrey MJ. The metabolism and pharmacokinetics of amlodipine in humans and animals. J Cardiovasc Pharmacol. 1988;12(Suppl 7):S55–9. doi: 10.1097/00005344-198812007-00012. [PubMed] [CrossRef] [Google Scholar]
31. Küng K, Wanner M. Bioavailability of different forms of amoxycillin administered orally to dogs. Vet Rec. 1994;135(23):552–4. [PubMed] [Google Scholar]
32. McAinsh J, Holmes BF. Pharmacokinetic studies with atenolol in the dog. Biopharm Drug Dispos. 1983;4(3):249–61. doi: 10.1002/bdd.2510040306. [PubMed] [CrossRef] [Google Scholar]
33. Shepard RM, Falkner FC. Pharmacokinetics of azithromycin in rats and dogs. J Antimicrob Chemother. 1990;25(Suppl A):49–60. doi: 10.1093/jac/25.suppl_A.49. [PubMed] [CrossRef] [Google Scholar]
34. Beddies G, Fox PR, Papich MG, Kanikanti VR, Krebber R, Keene BW. Comparison of the pharmacokinetic properties of bisoprolol and carvedilol in healthy dogs. Am J Vet Res. 2008;69(12):1659–63. doi: 10.2460/ajvr.69.12.1659. [PubMed] [CrossRef] [Google Scholar]
35. Carli S, Anfossi P, Villa R, Castellani G, Mengozzi G, Montesissa C. Absorption kinetics and bioavailability of cephalexin in the dog after oral and intramuscular administration. J Vet Pharmacol Ther. 1999;22(5):308–13. doi: 10.1046/j.1365-2885.1999.00208.x. [PubMed] [CrossRef] [Google Scholar]
36. Athanikar NK, Chiou WL. Chlorpheniramine. II. Effect of the first-pass metabolism on the oral bioavailability in dogs. J Pharmacokinet Biopharm. 1979;7(4):383–96. doi: 10.1007/BF01062536. [PubMed] [CrossRef] [Google Scholar]
37. Jantratid E, Prakongpan S, Dressman JB, Amidon GL, Junginger HE, Midha KK, et al. Biowaiver monographs for immediate release solid oral dosage forms: cimetidine. J Pharm Sci. 2006;95(5):974–84. doi: 10.1002/jps.20614. [PubMed] [CrossRef] [Google Scholar]
38. Le Traon G, Burgaud S, Horspool LJI. Pharmacokinetics of cimetidine in dogs after oral administration of cimetidine tablets. J Vet Pharmacol Ther. 2008;32:213–8. doi: 10.1111/j.1365-2885.2008.01026.x. [PubMed] [CrossRef] [Google Scholar]
39. Papich MG. Ciprofloxacin pharmacokinetics and oral absorption of generic ciprofloxacin tablets in dogs. Am J Vet Res. 2012;73(7):1085–91. doi: 10.2460/ajvr.73.7.1085. [PubMed] [CrossRef] [Google Scholar]
40. Batzias GC, Delis GA, Athanasiou LV. Clindamycin bioavailability and pharmacokinetics following oral administration of clindamycin hydrochloride capsules in dogs. Vet J. 2005;179:339–45. doi: 10.1016/j.tvjl.2004.06.007. [PubMed] [CrossRef] [Google Scholar]
41. King JN, Maurer MP, Holz RP, Fisch RD. Pharmacokinetics of clomipramine in dogs following single-dose intravenous and oral administration. Am J Vet Res. 2000;61:74–9. doi: 10.2460/ajvr.2000.61.74. [PubMed] [CrossRef] [Google Scholar]
42. KuKanich B. Pharmacokinetics of acetaminophen, codeine, and the codeine metabolites morphine and codeine-6-glucuronide in healthy Greyhound dogs. J Veterinary Pharmacol Ther. 2009;33:15–21. doi: 10.1111/j.1365-2885.2009.01098.x.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
43. Novartis Report. Pharmacokinetics of cyclosporine A after intravenous and oral administration to dogs. Novartis internal report, Study CRA 99/141, 2000.
44. Löscher W, Frey H-H. Pharmacokinetics of diazepam in the dog. Arch Int Pharmacodyn Ther. 1981;254:180–95. [PubMed] [Google Scholar]
45. Yeung PK, Feng JD, Buckley SJ. Effect of administration route and length of exposure on pharmacokinetics and metabolism of diltiazem in dogs. Drug Metabol Drug Interact. 2001;18(3–4):251–62. [PubMed] [Google Scholar]
46. Button C, Gross DR, Johnston JT, Yakatan GJ. Pharmacokinetics, bioavailability, and dosage regimens of digoxin in dogs. Am J Vet Res. 1980;41(8):1230–7. [PubMed] [Google Scholar]
47. Dow J, Francesco GF, Berg C. Comparison of the pharmacokinetics of dolasetron and its major active metabolite, reduced dolasetron, in dog. J Pharm Sci. 1996;85(7):685–9. doi: 10.1021/js960041m. [PubMed] [CrossRef] [Google Scholar]
48. Jantratid E, Strauch S, Becker C, Dressman JB, Amidon GL, Junginger HE, et al. Biowaiver monographs for immediate release solid oral dosage forms: doxycycline hyclate. J Pharm Sci. 2010;99(4):1639–53. [PubMed] [Google Scholar]
49. Fluoxetine. Manufacturer’s label. Reconcile® (fluoxetine hydrochloride) chewable tablets. INADA # 141–272. Elanco Animal Health, Indianapolis, IN, USA.
50. El-Sayed MG, Atef M, El-Gendi AY, Youssef SA. Disposition kinetics of furosemide in dogs. Arch Int Pharmacodyn Ther. 1981;253(1):4–10. [PubMed] [Google Scholar]
51. Lee MG, Chiou WL. Mechanism of ascorbic acid enhancement of the bioavailability and diuretic effect of furosemide. Drug Metab Dispos. 1998;26(5):401–7. [PubMed] [Google Scholar]
52. Radulovic LL, Türck D, von Hodenberg A, Vollmer KO, McNally WP, DeHart PD, et al. Disposition of gabapentin (neurontin) in mice, rats, dogs, and monkeys. Drug Metab Dispos. 1995;23(4):441–8. [PubMed] [Google Scholar]
53. Semple HA, Tam YK, Coutts RT. Hydralazine pharmacokinetics and interaction with food: an evaluation of the dog as an animal model. Pharm Res. 1990;7(3):274–9. doi: 10.1023/A:1015830330231. [PubMed] [CrossRef] [Google Scholar]
54. Bizikova P, Papich MG, Olivry T. Hydroxyzine and cetirizine pharmacokinetics and pharmacodynamics after oral and intravenous administration of hydroxyzine to healthy dogs. Vet Dermatol. 2008;19(6):348–57. doi: 10.1111/j.1365-3164.2008.00697.x. [PubMed] [CrossRef] [Google Scholar]
55. Scherkl R, Frey H-H. Pharmacokinetics of ibuprofen in the dog. J Vet Pharmacol Ther. 1987;10:261–5. doi: 10.1111/j.1365-2885.1987.tb00539.x. [PubMed] [CrossRef] [Google Scholar]
56. Schmitt M, Guentert TW. Biopharmaceutical evaluation of ketoprofen following intravenous, oral, and rectal administration in dogs. J Pharma Sci. 1990;79:614–6. doi: 10.1002/jps.2600790715. [PubMed] [CrossRef] [Google Scholar]
57. Moore SA, Muñana KR, Papich MG, Nettifee-Osborne J. Levetiracetam pharmacokinetics in healthy dogs following oral administration of single and multiple doses. Am J Vet Res. 2010;71(3):337–41. doi: 10.2460/ajvr.71.3.337. [PubMed] [CrossRef] [Google Scholar]
58. Slatter JG, Stalker DJ, Feenstra KL, Welshman IR, Bruss JB, Sams JP, et al. Pharmacokinetics, metabolism, and excretion of linezolid following an oral dose of [(14)C]linezolid to healthy human subjects. Drug Metab Dispos. 2001;29(8):1136–45. [PubMed] [Google Scholar]
59. Slatter JG, Adams LA, Bush EC, Chiba K, Daley-Yates PT, Feenstra KL, et al. Pharmacokinetics, toxicokinetics, distribution, metabolism, and excretion of linezolid in mouse, rat, and dog. Xenobiotica. 2002;32:907–24. doi: 10.1080/00498250210158249. [PubMed] [CrossRef] [Google Scholar]
60. Busch U, Schmid J, Heinzel G, Schmaus H, Baierl J, Huber C, et al. Pharmacokinetics of meloxicam in animals and the relevance to humans. Drug Metab Dispos. 1998;26(6):576–84. [PubMed] [Google Scholar]
61. Bateman DN, Kahn C, Davies DS. The pharmacokinetics of metoclopramide in man with observations in the dog. Br J Clin Pharmacol. 1980;9:371–7. doi: 10.1111/j.1365-2125.1980.tb01064.x.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
62. Neff-Davis CA, Davis LE, Gillette EL. Metronidazole: a method for its determination in biological fluids and its disposition kinetics in the dog. J Vet Pharmacol Ther. 1981;4(2):121–7. doi: 10.1111/j.1365-2885.1981.tb00720.x. [PubMed] [CrossRef] [Google Scholar]
63. Maaland MG, Guardabassi L, Papich MG. Minocycline pharmacokinetics and pharmacodynamics in dogs: dosage recommendations for treatment of meticillin‐resistant Staphylococcus pseudintermedius infections. Vet Dermatol. 2014;25(3):182. doi: 10.1111/vde.12130. [PubMed] [CrossRef] [Google Scholar]
64. KuKanich B, Lascelles BD, Papich MG. Pharmacokinetics of morphine and plasma concentrations of morphine-6-glucuronide following morphine administration to dogs. J Vet Pharmacol Ther. 2005;28(4):371–6. doi: 10.1111/j.1365-2885.2005.00661.x. [PubMed] [CrossRef] [Google Scholar]
65. Frey H-H, Rieh B. Pharmacokinetics of naproxen in the dog. Am J Vet Res. 1981;42:1615–7. [PubMed] [Google Scholar]
66. Pedersoli WM, Wike JS, Ravis WR. Pharmacokinetics of single doses of phenobarbital given intravenously and orally to dogs. Am J Vet Res. 1987;48:679–83. [PubMed] [Google Scholar]
67. Frey H-H, Löscher W. Clinical pharmacokinetics of phenytoin in the dog: a reevaluation. Am J Vet Res. 1980;41(10):1635–8. [PubMed] [Google Scholar]
68. Galbraith EA, McKellar QA. Pharmacokinetics and pharmacodynamics of piroxicam in dogs. Vet Rec. 1991;128:561–5. doi: 10.1136/vr.128.24.561. [PubMed] [CrossRef] [Google Scholar]
Bcs Classification Database
69. Papich MG, Neff-Davis CA, Davis LE, McKiernan BC, Brown SA. Pharmacokinetics of procainamide in dogs. Am J Vet Res. 1986;47:2351–8. [PubMed] [Google Scholar]
70. Tse FL, Sanders TM, Reo JP. Bioavailability of propranolol in the dog. Arch Int Pharmacodyn Ther. 1980;248(2):180–9. [PubMed] [Google Scholar]
71. Halpin RA, Geer LA, Zhang KE, Marks TM, Dean DC, Jones AN, et al. The absorption, distribution, metabolism and excretion of rofecoxib, a potent and selective cyclooxygenase-2 inhibitor, in rats and dogs. Drug Metab Dispos. 2000;28(10):1244–54. [PubMed] [Google Scholar]
72. Walker DK, Ackland MJ, James GC, Muirhead GJ, Rance DJ, Wastall P, et al. Pharmacokinetics and metabolism of sildenafil in mouse, rat, rabbit, dog and man. Xenobiotica. 1999;29(3):297–310. doi: 10.1080/004982599238687. [PubMed] [CrossRef] [Google Scholar]
73. McKiernan BC, Neff-Davis CA, Koritz GD, Davis LE, Pheris DR. Pharmacokinetic studies of theophylline in dogs. J Vet Pharmacol Ther. 1981;4:103–10. doi: 10.1111/j.1365-2885.1981.tb00718.x. [PubMed] [CrossRef] [Google Scholar]
74. Bach JE, Kukanich B, Papich MG, McKiernan BC. Evaluation of the bioavailability and pharmacokinetics of two extended-release theophylline formulations in dogs. J Am Vet Med Assoc. 2004;224(7):1113–9. doi: 10.2460/javma.2004.224.1113. [PubMed] [CrossRef] [Google Scholar]
75. KuKanich B, Papich MG. Pharmacokinetics and antinociceptive effects of oral tramadol hydrochloride administration in Greyhounds. Am J Vet Res. 2011;72(2):256–62. doi: 10.2460/ajvr.72.2.256. [PubMed] [CrossRef] [Google Scholar]
76. Benet LZ, Broccatelli F, Oprea TI. BDDCS applied to over 900 drugs. AAPS J. 2011;13(4):519–47. doi: 10.1208/s12248-011-9290-9.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
77. Arndt M, Chokshi H, Tang K, Parrott NJ, Reppas C, Dressman JB. Dissolution media simulating the proximal canine gastrointestinal tract in the fasted state. Eur J Pharm Biopharm. 2013;84(3):633–41. doi: 10.1016/j.ejpb.2013.01.010. [PubMed] [CrossRef] [Google Scholar]
78. Dressman JB, Reppas C. In vitro-in vivo correlations for lipophilic, poorly water-soluble drugs. Eur J Pharm Sci Suppl. 2000;2:S73–80. doi: 10.1016/S0928-0987(00)00181-0. [PubMed] [CrossRef] [Google Scholar]
79. Bretschneider B, Brandsch M, Neubert R. Intestinal transport of beta-lactam antibiotics: analysis of the affinity at the H+/peptide symporter (PEPT1), the uptake into Caco-2 cell monolayers and the transepithelial flux. Pharm Res. 1999;16(1):55–61. doi: 10.1023/A:1018814627484. [PubMed] [CrossRef] [Google Scholar]
80. Ranaldi G, Islam K, Sambuy Y. D-cycloserine uses an active transport mechanism in the human intestinal cell line Caco 2. Antimicrob Agents Chemother. 1994;38(6):1239–45. doi: 10.1128/AAC.38.6.1239.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
81. Westphal JF, Jehl F, Brogard JM, Carbon C. Amoxicillin intestinal absorption reduction by amiloride: possible role of the Na(+)-H+ exchanger. Clin Pharmacol Ther. 1995;57(3):257–64. doi: 10.1016/0009-9236(95)90150-7. [PubMed] [CrossRef] [Google Scholar]
82. Shugarts S, Benet LZ. The role of transporters in the pharmacokinetics of orally administered drugs. Pharm Res. 2009;26:2039–54. doi: 10.1007/s11095-009-9924-0.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
83. Flanagan SD, Takahashi LH, Liu X, Benet LZ. Contributions of saturable active secretion, passive transcellular, and paracellular diffusion to the overall transport of furosemide across adenocarcinoma (Caco-2) cells. J Pharm Sci. 2002;91(4):1169–77. doi: 10.1002/jps.10099. [PubMed] [CrossRef] [Google Scholar]
84. Flanagan SD, Cummins CL, Susanto M, Liu X, Takahashi LH, Benet LZ. Comparison of furosemide and vinblastine secretion from cell lines overexpressing multidrug resistance protein (P-glycoprotein) and multidrug resistance-associated proteins (MRP1 and MRP2) Pharmacology. 2002;64(3):126–34. doi: 10.1159/000056161. [PubMed] [CrossRef] [Google Scholar]
85. Al-Mohizea AM. Influence of intestinal efflux pumps on the absorption and transport of furosemide. Saudi Pharm J. 2010;18(2):97–101. doi: 10.1016/j.jsps.2010.02.005.[PMC free article] [PubMed] [CrossRef] [Google Scholar]
86. Kukanich B, Papich MG. Plasma profile and pharmacokinetics of dextromethorphan after intravenous and oral administration in healthy dogs. J Vet Pharmacol Ther. 2004;27(5):337–41. doi: 10.1111/j.1365-2885.2004.00608.x. [PubMed] [CrossRef] [Google Scholar]
87. Kukanich B, Lascelles BD, Aman AM, Mealey KL, Papich MG. The effects of inhibiting cytochrome P450 3A, p-glycoprotein, and gastric acid secretion on the oral bioavailability of methadone in dogs. J Vet Pharmacol Ther. 2005;28(5):461–6. doi: 10.1111/j.1365-2885.2005.00681.x. [PubMed] [CrossRef] [Google Scholar]
88. Ingels F, Beck B, Oth M, Augustijns P. Effect of simulated intestinal fluid on drug permeability estimation across Caco-2 monolayers. Int J Pharm. 2004;274(1–2):221–32. doi: 10.1016/j.ijpharm.2004.01.014. [PubMed] [CrossRef] [Google Scholar]
89. Miller JM, Beig A, Krieg BJ, Carr RA, Borchardt TB, Amidon GE, et al. The solubility-permeability interplay: mechanistic modeling and predictive application of the impact of micellar solubilization on intestinal permeation. Mol Pharm. 2011;8(5):1848–56. doi: 10.1021/mp200181v. [PubMed] [CrossRef] [Google Scholar]
90. Markopoulos C, Imanidis G, Vertzoni M, Symillides M, Parrott N, Reppas C. In vitro and ex vivo investigation of the impact of luminal lipid phases on passive permeability of lipophilic small molecules using PAMPA. Pharm Res. 2013;30(12):3145–53. doi: 10.1007/s11095-013-1141-1. [PubMed] [CrossRef] [Google Scholar]
91. Brouwers J, Augustijns P. Resolving intraluminal drug and formulation behavior: gastrointestinal concentration profiling in humans. Eur J Pharm Sci. 2014 [PubMed] [Google Scholar]
92. Dressman JB. Comparison of canine and human gastrointestinal physiology. Pharm Res. 1986;3:123–31. doi: 10.1023/A:1016353705970. [PubMed] [CrossRef] [Google Scholar]
93. Wynalda MA, Hutzler JM, Koets MD, Podoll T, Wienkers LC. In vitro metabolism of clindamycin in human liver and intestinal microsomes. Drug Metab Disp. 2003;31(7):878--887. [PubMed]
94. Martinez MN, Papich MG. Factors influencing the gastric residence of dosage forms in dogs. J Pharm Sci. 2009;98(3):844–60. doi: 10.1002/jps.21499. [PubMed] [CrossRef] [Google Scholar]
95. Kararli TT. Comparison of the gastrointestinal anatomy, physiology, and biochemistry of humans and commonly used laboratory animals. Biopharm Drug Dispos. 1995;16:351–80. doi: 10.1002/bdd.2510160502. [PubMed] [CrossRef] [Google Scholar]
96. Sabnis S. Factors influencing the bioavailability of peroral formulations of drugs for dogs. Vet Res Commun. 1999;23:425–47. doi: 10.1023/A:1006321625243. [PubMed] [CrossRef] [Google Scholar]
97. Kolars JC, Awni WM, Merion RM, Watkins PB. First pass metabolism of cyclosporin by the gut. Lancet. 1991;338(8781):1488–90. doi: 10.1016/0140-6736(91)92302-I. [PubMed] [CrossRef] [Google Scholar]
98. Oliyai R, Stella VJ. Kinetics and mechanism of isomerization of cyclosporin A. Pharm Res. 1992;9(5):617–22. doi: 10.1023/A:1015841824760. [PubMed] [CrossRef] [Google Scholar]
99. Piyapolrungroj N, Li C, Pisoni RL, Fleisher D. Cimetidine transport in brush-border membrane vesicles from rat small intestine. JPET. 1999;289(1):346--353. [PubMed]
100. Musther H, Olivares-Morales A, Hatley OJD, Liu B, Hodjegan AR. Animal versus human oral bioavailability: Do they correlate? Eur J Pharm Sci. (2014);57: 280–291. [PMC free article] [PubMed]
Articles from The AAPS Journal are provided here courtesy of American Association of Pharmaceutical Scientists

Services on Demand

  • Cited by SciELO

Related links

  • Similars in SciELO

On-line version ISSN 2175-9790

http://dx.doi.org/10.1590/S1984-82502015000100015

Articles

Biopharmaceutics classification system: importance and inclusion in biowaiver guidance

1Universidade Federal de Ouro Preto, Escola de Farmácia, Departamento de Farmácia, Ouro Preto, MG, Brazil

Pharmacological therapy is essential in many diseases treatment and it is important that the medicine policy is intended to offering safe and effective treatment with affordable price to the population. One way to achieve this is through biowaiver, defined as the replacement of in vivo bioequivalence studies by in vitro studies. For biowaiver of new immediate release solid oral dosage forms, data such as intestinal permeability and solubility of the drug are required, as well as the product dissolution. The Biopharmaceutics Classification System (BCS) is a scientific scheme that divides drugs according to their solubility and permeability and has been used by various guides as a criterion for biowaiver. This paper evaluates biowaiver application, addressing the general concepts and parameters used by BCS, making a historical account of its use, the requirements pertaining to the current legislation, the benefits and risks associated with this decision. The results revealed that the use of BCS as a biowaiver criterion greatly expands the therapeutics options, contributing to greater therapy access of the general population with drug efficacy and safety guaranteed associated to low cost.

Key words: Medicines/biowaiver; Biopharmaceutics Classification System; Drugs/solubility; Drugs/permeability; Drugs/dissolution; Drugs/legislation

O tratamento farmacológico é essencial frente a várias patologias e é fundamental que a política de medicamentos tenha por objetivo oferecer à população tratamento seguro, eficaz e de preço acessível. Uma forma de alcançar esse objetivo é por meio da bioisenção, definida como a substituição de estudos de bioequivalência in vivo por estudos in vitro. Para bioisentar novos medicamentos sob a forma farmacêutica sólida oral de liberação imediata são utilizados dados de permeabilidade intestinal e solubilidade do fármaco, bem como sua dissolução a partir da forma farmacêutica. O Sistema de Classificação Biofarmacêutica (SCB) é um esquema científico que divide os fármacos em classes de acordo com a solubilidade e permeabilidade e vem sendo utilizado como critério para bioisenção em diversas legislações. O presente artigo faz uma avaliação da aplicação da bioisenção, abordando os conceitos gerais e parâmetros utilizados pelo SCB, fazendo um relato histórico da aplicação da bioisenção, das exigências pertinentes às legislações vigentes, dos benefícios e riscos inerentes a uma tomada de decisão sobre bioisenção baseada neste critério. Os resultados revelaram que a utilização do SCB como critério amplia enormemente as possibilidades de bioisenção, contribuindo para o maior acesso da população em geral a medicamentos com garantida eficácia, segurança e menor custo.

Palavras-Chave: Medicamentos/bioisenção; Sistema de Classificação Biofarmacêutica; Fármaco/solubilidade; Fármaco/permeabilidade; Fármaco/dissolução; Fármaco/legislação

INTRODUCTION

Medicines are the most important tool that society possesses to prevent, alleviate and cure diseases (Leach, Palluzi, Munderi, 2005). However, about 30% of the world population has no access to effective, safe and quality medicines,and more than half of those live in developing countries in Africa and Asia (WHO, 2004).This problem is so serious that in 2000 the United Nations (UN), analyzing the major problems of the world, established the Millennium Program, where 191 countries signed an agreement with 8 goals and 18 targets that must be achieved until 2015. One of these goals is to provide access to quality essential medicines at affordable prices (Brasil, 2007).

Several factors influence people's access to medicines and it is necessary a national policy to regulate and monitor these several factors. Among those we may cite: a system for efficient drug selection and free from pressures, a policy of public funded sustainable system for regulating the drug market, controlling overtaxes and other factors that impact on prices, as well as an efficient distribution system (Leach, Palluzi, Munderi, 2005).

Ge smart connection center panel. In this sense, the policy of generic medicines is an important strategy for increasing access to essential medicines, particularly in developing countries by reducing costs, both for the consumer as for purchase by the public system, for subsequent distribution to the population.

Some defined dosage forms of generic medicines must necessarily prove its bioequivalence in relation to the reference product (Miranda et al., 2009). This testing assess the intensity and extent of absorption in humans and to do this, the costs are high, inherent to the clinical, analytical and statistical steps that consume the time of 3 to 7 months between the selection of which center will elaborate the studies, approval by the ethics committee on research, development and selection of volunteers for those steps. However, this provides consumers with the opportunity to purchase medicines at more affordable prices, with a guarantee of quality, safety, efficiency and interchangeability with the reference medicines (Araújo et al., 2010; Melo, 2005).

The Food and Drug Administration (FDA), the U.S. regulatory agency, estimates that in coming years, US$ 60 to US$ 70 billion in branded medicines will lose their patent protection (United States, 2007). This means that these drugs may be considered to be registered as generic, but for this, it is necessary knowledge and structure able to respond quickly to a potential increase in demand for generic drugs registration. In order to do that, biowaiver can be a good tool and thus bring benefits to the society by expanding the supply of medicines effective with guaranteed security. The biowaiver can be defined as the acceptance for regulatory purposes, of the exemption or replacement of in vivo bioequivalence studies and bioavailability by in vitro assays when they are able to replace the in vivo assay reliably (Storpirtis, Gai, 2009). The registration process through biowaiver enables a reduction in the exposure of volunteers in clinical studies, time and cost of product development with quality and safety certified (Lennernäs, Abrahamsson, 2005). Therefore, the biowaiver brings considerable benefits for the patient and government health systems. In Brazil, it is an important contribution to the national health system (SUS) in terms of access to drug treatment. The Food and Drug Administration and Agência Nacional de Vigilância Sanitária (ANVISA) that is the Brazilian regulatory agency, use the Biopharmaceutics Classification System (BCS) as a mean to allow biowaiver of Immediate-Release Solid Oral Dosage Forms containing drugs with high solubility and permeability (BCS Class I) (United States, 2000Brasil, 2011). The European Union agency (EMA) responsible to indicate candidates to biowaiver has considered eligible the class I of BCS and additionally, drugs which have high solubility and low permeability (BCS Class III), when they show very rapid dissolving (85% in 15 minutes). The World Health Organization (WHO) further extends the applicability of biowaiver for drugs in classes I, III and also drugs poorly soluble and highly permeable (Class II), when they have characteristics of weak acid and high solubility in pH 6,8, besides rapid or very rapid dissolving (EMEA, 2008; WHO, 2006).

This article aims to contribute to the understanding of the general concepts on the topic and discuss the inclusion of BCS as a biowaiver criterion. Moreover, it presents the historical account of the relevant legislation, effective criteria to apply biowaiver and discusses the benefits and risks of using the BCS as a regulatory tool.

METHODS

The laws related to biowaiver were checked at sites of the regulatory agencies of interest, ANVISA (http://www.anvisa.gov.br/e-legis), FDA (http://www.fda.gov/default.htm) and EMA (http://www.ema.europa.eu/ema/), to evaluate the inclusion of BCS as a criterion to enable biowaiver. Additionally, we carried out search of scientific basis for allowing the presentation and discussion of the laws cited. With this goal bibliographic data bases PubMed (www.ncbi.nlm.nih.gov), Scopus (www.scopus.com/home.url) and SciELO (www.scielo.org) were consulted from November 2011 to February 2014, using the following search terms: biowaiver, BCS, permeability and solubility.

THEORETICAL BASIS FOR BIOWAIVER

Biopharmaceutical classification system

The Biopharmaceutical Classification System is a scientific schematic proposed by Amidon and coworkers in 1995, which allowed the classification of drugs based on solubility and intestinal permeability parameters in four classes (Amidon et al., 1995).

The four possible categories for a drug according to the BCS are in Table I

Table 1. The Biopharmaceutical Classification System scientific framework

Class Solubility Permeability
I High High
II Low High
III High Low
IV Low Low

The solubility and intestinal permeability are among the main factors that govern the rate and extent of drug absorption and therefore are directly related to bioavailability (Amidon et al., 1995).

The insertion of the BCS as a criterion for biowaiver started in FDA guide, also being currently accepted by ANVISA and EMA. This addition allowed the simplification of registration of new medicines according to Abbreviated New Drug Application (ANDA) in USA (Löbenberg, Amidon, 2000).

Solubility

By definition, solubility is the extent to which one molecule of a solid is removed from its surface by a solvent (Martinez, Amidon, 2002) being a determining factor for the absorption and bioavailability of active compounds (Panchagnula, Thomas, 2000).

A drug is considered highly soluble when the highest dose administered as an immediate release formulation, is soluble in 250 ml or less of aqueous media with a pH in a range of 1.2 to 6.8 at 37±1 °C (Brasil, 2011).

Permeability

The permeability is a dynamic and complex process that involves the permeation of the drug across biological membranes. The arrival of the drug into the bloodstream is made by the absorption pharmacokinetics processes. The transport mechanisms include passive diffusion through the enterocytes (transcellular) and the junctions between the enterocytes (paracellular) as well as active mechanisms employing energy and carriers (Balimane et al., 2000).

A drug is considered highly permeable when the extent of absorption in humans is 85% (Brasil, 2011; EMEA, 2008; WHO, 2006) or more, based on mass balance determination or in comparison with an intravenous dose. On the other hand, according to FDA (2000) one drug is classified as highly permeable when the fraction of the absorbed dose or the absolute bioavailability is equal or greater than 90%. This criterion can be considered conservative because there are many reports of drugs that are generally considered well or completely absorbed, that present fraction of absorbed dose less than 90%. This suggests that a threshold rating of 85% may be appropriate in the definition of high permeability (Yu et al., 2002).

The need to obtain data relating to intestinal permeability of drugs has raised the development of various models for the determination of permeability, which may be in vivo, in situ, in vitro and in silico (Souza, Freitas, Storpirtis, 2007). The methods continue to be improved and it is recommended that conclusions should be obtained by using more than one method (Balimane et al., 2000).

Bcs Classification System Database

Dissolution

The dissolution can be defined in a narrow sense as the process by which a solid substance enters the solvent to form a solution. However, in the broad sense of the word, it is more than simply measuring the rate of solubility and can be more correctly described as a physical assay to predict the release of substance for a given area, in quantity and adequate time (Manadas, Pina, Veiga, 2002).

The absorption of a drug contained in solid dosage form after oral administration depends on its release, solubilization and dissolution under physiological conditions and posterior permeability through the gastrointestinal tract. The first two steps present a critical nature and therefore, in vitro dissolution may be relevant to predict the in vivo performance. In vitro dissolution tests for oral solid dosage forms such as tablets and capsules are used to: [1] assess the quality of a batch to batch drug; [2] guide development of new formulations; [3] guarantee the maintenance of product quality after certain changes in the design, process, and on site production scale (United States, 1997) and [4] assess the pharmaceutical equivalence between products from different manufacturers (Rodrigues et al., 2006).

One way to evaluate the dissolution is by determining the dissolution profile, graph of percentage of drug dissolved versus time, which is a relatively quick and inexpensive way to evaluate solid dosage forms and allows obtaining kinetic parameters. These are important for determining the speed and efficiency of the process and the time needed for specific release of drug percentages, allowing conclusions about in vitro biopharmaceutical characteristics of the formulation (Storpirtis et al., 1999).

Pharmaceutical Equivalence

Pharmaceutical equivalence between two medicines is related to the confirmation that both contain the same drug (same basis, salt or ester of the same therapeutically active molecule) at the same dose and dosage form, and may be evaluated through in vitro tests (Storpirtis et al., 2004). To be considered pharmaceutical equivalents, the test and reference medicine must comply in full with the requirements of pharmacopoeia monographs, completed with assays described in general methods for the pharmaceutical form (Brasil, 2010).

Bioequivalence/Relative Bioavailability

Bioequivalence or relative bioavailability is the comparative study of products containing the same drug administered by the same route. Two products are considered bioequivalent if, when administered to the same individual under the same experimental conditions and at the same molar dose do not differ significantly from the amount of drug absorbed and the rate of absorption process (Storpirtis, Consiglieri, 1995).

Brazilian law states that for a drug to be registered as generic, it is necessary to prove their pharmaceutical equivalence and bioequivalence in respect to the reference product indicated by ANVISA (Brasil, 2003a). As for similar drugs products, it is required to prove pharmaceutical equivalence and similar relative bioavailability to the inclusion of a new product or renewal of registration of drugs already on the market (Brasil, 2003b).

For the registration of similar medicines in Brazil, up to the year 2003, it was not necessary the proof of bioavailability. According to RDC N.134/2003 the new reality of achieving this requirement must occur until 2014. For this process, ANVISA adopted 'Relative bioavailability' for similar medicines as a term to differentiate them from generic medicines (interchangeable), since the term 'bioequivalence' is established and accepted internationally for generic products (Araújo et al., 2010).

BIOWAIVER

A Biowaiver means that relative bioavailability and/or bioequivalence tests are not required for the registration of a medicine by the regulatory authority, when a suitable in vitro assay can replace it (Brasil, 2011).

The main reason for biowaiver based on the BCS is that in some situations, in vitro assays are as good as in vivo tests to determine the bioequivalence of oral solid dosage forms and sometimes better in terms of direct evaluation of product performance. Moreover, biowaiver eliminates unnecessary exposure of healthy subjects to in vivo studies, reduces the burden of evaluating petitions for registration requiring BE studies, and provides economic relief, maintaining the quality standard of dispensed medicines to public health and thus ensuring therapeutic equivalence (Cook et al., 2010).

Bcs

An estimated 66 to 76 million dollars can be saved each year in costs of clinical studies if biowaiver is granted to all applications for new drugs containing drug class I. If this is extended to class III compounds, additional savings from 62 to 71 million dollars can be performed (Cook et al., 2010).

BCS AS BIOWAIVER CRITERION

The first legislation of biowaiver which includes the BCS as a criterion and that is still in use, was published by the FDA in August 2000. This exemption would be based on the grounds that, if two formulations present the same dissolution profile in vivo, under the same conditions of the intestinal lumen, they present the same profile of concentration versus time on the surface of the intestinal membrane, which results in the same rate and extent of absorption (United States, 2000).

Bcs Classification Database Fda

ANVISA, as well as FDA, in its recently published guide, RDC Resolution No. 37, August 3, 2011 (Brazil, 2011), recommends the exemption of bioavailability/ bioequivalence studies in cases of drugs highly soluble and highly permeable (Class I) into oral solid dosage forms that provide rapid dissolving, in other words, not less than 85% in 30 minutes. Moreover, evidence of solubility, permeability and dissolution should be performed according to the methods proposed in the legislation (United States, 2007; Brasil, 2011).The normative statement (IN) No.2, (ANVISA, 2013) is a document that refers to a list of drugs whose biopharmaceutical characteristics were established and are suitable to pass by the biowaiver process according to RDC Resolution No. 37, August 3, 2011.

In Europe, the EMA published in 2001 its biowaiver guide, which was updated in 2008. In this resolution, FDA and ANVISA recommend the BCS biowaiver in similar way, i.e. for medicines containing drugs class I. Besides these, EMA also considers the biowaiver based on the BCS for class III drugs, i.e. the medicines which have high solubility and limited absorption and also have very fast in vitro dissolution characteristics (85% in 15 minutes), excipients qualitatively and quantitatively identical (EMEA, 2008; EMEA, 2001).

In 2006, the World Health Organization (WHO) published a document entitled 'WHO Technical Report Series No. 937' that has an attachment about biowaiver theme ('Proposal to waive in vivo bioequivalence requirements for WHO Model List of Essential Medicines immediate-release, solid oral dosage forms', Annex 8). This guide recommends criteria of biowaiver based on BCS, however, its requirements are less stringent than those set by the guides of the FDA, ANVISA and EMA, including drugs of class II, low solubility and high permeability, as liable to biowaiver (when they have characteristics of weak acid and high solubility in pH 6,8, besides rapid or very rapid dissolving). The WHO recommendation is to establish less conservative criteria, especially for well-known drugs, such as those that are part of the List of Essential Medicines (WHO, 2006).

In short, according to present guides, for a drug to have its biowaiver accepted, it must have its biopharmaceutical classification proven, according to the criteria of solubility and permeability, as well as dissolution characteristics from the dosage form. Table II has a comparison of the main requirements for obtaining biowaiver based on the BCS, according to regulatory agencies ANVISA, FDA, EMA and WHO. In addition, to meet these criteria, an analysis of the formulation of the biowaiver candidate should also be performed as well as a parsing of the risk of an incorrect decision for individual and collective health (WHO, 2006; Cook, Addicks, Wu, 2008).

Table 2. Main requirements for obtaining biowaiver based on BCS

REGULATORYAGENCY
FDA ANVISA EMA OMS
Candidates according to BCS Class I Class I and III** Class I, II* e III**
Characteristic for high solubility classification of drugs Higher dose commercially available must be soluble in ≤ 250 mL, pH 1-7.5,
37 °C
Highest single dose administered to the patient and provided in package insert must be soluble in ≤ 250 mL,pH 1.2 to 6.8, 37 ⁰C.
Classification of high permeability drugs Superior to metoprolol absorption (absorption≥ 90 % of administered dose) Absorption ≥85%
Dissolution media buffer pH1.2, 4.5 and 6.8; 37 °C
Volume of medium 900 mL 500 mL 900 mL
Apparatus and rotation Paddle: 50 rpm/Basket: 100 rpm. Paddle: 75 rpm/
Basket: 100 rpm.
Dissolution profile I:Rapid dissolving I: Rapid dissolving
III: Very rapid
dissolving
I: Rapid dissolving
II: Rapid dissolving
at pH 6,8
III: Very rapid dissolving
Recommendations about the excipient Excipients used in the dosage form must have been used in a previously approved immediate release (IR) solid oral dosage form by the Food and Drug Administration . Qualitatively the same and quantitatively very similar to the respective reference drug product Absence of excipients that have an impact on bioavailability.

* Weak acid, high solubility in pH 6,8, rapid or very rapid dissolving. ** Very rapid dissolving (85% in 15 minutes)

The quantity of excipients in the IR product should be consistent with their intended function. Large quantities of certain excipients, such as surfactants (e.g., sodium laurylsulfate) or osmotic ingredients (e.g., sorbitol) may be problematic and should be avoided except when present in the reference drug (United States, 2000).

Since 1977, WHO has periodically prepared a list of essential medicines for basic health care. Two studies, one conducted by Kasim and colleagues, in 2004, and the other held by Lindenberg, Kopp, Dressman, in 2004, provisionally classified by the BCS, drugs presented at the 12th edition of this list (2002). The results of these studies have been compiled and presented in Figure 1.

Figure 1. Provisional Classification of drugs present in the 12th edition of the essential drugs list proposed by Kasim et al. (2004) and Lindenberg, Kopp, Dressman (2004) .

In 2006, Takagi and colleagues (2006) also classified by the BCS the 200 best-selling drugs in the United States, Spain, Britain and Japan. In short, over 55% of these drugs were classified as highly soluble (class I and III), and approximately 30% of all drugs contained in oral solid dosage forms may be classified as highly soluble and highly permeable (Class I).

One of the main initiatives of the FDA regarding biowaiver based on BCS was to form a Technical Committee to review the requests of biowaiver in March of 2004. This committee met six times between 2004 to 2006 and evaluated 25 requests of biowaiver, 11 for new molecules and 14 for generics. Of the 11 requests for new molecules analyzed, 7 of them were for drugs of class I and had biowaiver accepted. Of the 14 requests for generics biowaiver, 9 drugs belonged to class I. Of these, four were accepted (Polli et al., 2008).

Biowaiver requests based on BCS were also analyzed by EMA, and the following have been accepted for biowaiver: phenoxymethylpenicillin, prednisolone, transexamico acid, acetaminophen, codeine and ibuprofen in Sweden (Graffner, 2006), and sotalol hydrochloride in Germany (Alt et al., 2004).

A new approach to ensure a safe decision about biowaiver initiated in 2004, by the International Pharmaceutical Federation (FIP) that has published a number of monographs containing relevant data available on literary sources for a given drug. These monographs are intended to support the discussion of risk associated with biowaiver of certain drugs selected by a group of experts from the FIP included in List of Essential Drugs. For this proposal, risk is defined as the probability of an incorrect biowaiver decision, as well as their consequences in terms of public health and risk to patients. This initiative has the support of WHO and aims especially to assist developing countries, in the approval and registration of generic medicines through biowaiver (FIP).

In these monographs, a recommendation was made about the possibility to biowaiver and although they do not have formal regulatory status, represent the best currently available scientific opinion. Until 2009, approximately 26 drugs have been evaluated in these monographs and the exemption of in vivo BE was scientifically justified and recommended for the vast majority of them. These monographs are part of an ongoing project, and the details and progress of this plan are available in www.fip.org/bcs (Dahan et al., 2009). So far, in 2014, around 40 monographs dealing with drugs have been written.

BIOWAIVER PETITIONS BASED ON BCS

Although there was an increase in the number of applications of biowaiver based on BCS, this progress has been tempered by the lack of international harmonization and the reluctance of companies to adhere to the methodology due to fears raised by a possible delay in the registers. Future progress in the use of biopharmaceutical in vitro data to substitute in vivo data by bioequivalence can also lead to benefits to achieve greater certainty in regulatory decisions, e.g. more scientific opportunities to discuss the necessary data and successful examples (Polli et al., 2008).

As previously mentioned, approximately 30% of all drugs contained in oral solid dosage forms may be classified as highly soluble and highly permeable (Takagi et al., 2006). So it may be surprising that, between 2003 and 2006, only 25 requests (11 for new drugs and 14 for generics) were submitted to the FDA for exemption from an in vitro study based on biopharmaceutical classification. The perceived lack of certainty of the acceptance by regulatory agencies of biowaiver requests based on BCS has been cited as one reason for the small number of requests by pharmaceutical industry (Polli et al., 2008).

Despite agencies like the FDA undertake significant efforts to promote the use of biowaiver based BCS, the time between the submission of an application for biowaiver and the formal approval needs to be fast enough (e.g., within weeks instead months), to have the adhesion of pharmaceutical companies. Not granting the biowaiver approval can lead to a significant delay in the development program of a new pharmaceutical product. Initially, the risk associated with receiving a negative response from biowaiver seemed greater than the benefits of economy of resources (Cook, Addicks, Wu, 2008). Nowadays, greater disclosure procedures for testing solubility and permeability as well as the expansion and dissemination of studies of drugs that could be waiver of bioequivalence studies contributes to expanding the adherence of the pharmaceutical industry (ANVISA, 2013; Cristofoletti et al., 2013).

Another barrier to the application of biowaiver refers to compartmentalization of some large companies, which can cause doubt as to the allocation of financial resources and the allocation of responsibilities regarding the success or failure of the biowaiver request. In the first case, the values saved by an organization resulting from a biowaiver typically appear in the clinical department budget. However, departments such as preclinical pharmacokinetic, chemistry and formulation may be required to perform more than the normal amount of work to support the application of a biowaiver. As a result, there may be reluctance by all parts of the organization to support a strategy for biowaiver. In the second case, the difficulty to employ biowaiver based on BCS can also be caused by lack of clarity on the responsibility to generate documents for biowaiver and to the attribution of blame, if a request of biowaiver is rejected and the time to launch a new product is delayed (Cook, Addicks, Wu, 2008).

DISCUSSION

The BCS developed by Amidon et al. (1995) aimed to employ specific tests in vitro in order to predict the drugs dissolution and therefore to estimate the results of their bioavailability (BD) in vivo, produced a significant impact on drug policy, making it possible to exempt BD and BE tests in vivo for class I drugs in oral solid dosage forms (FDA, EMA, ANVISA and WHO) for class III (EMA and WHO) and for a specific group of class II compounds (WHO). This is highly relevant for the possible decrease in the costs for registration of new generic drugs (Ramirez et al., 2010).

However, as shown in Figure 1, there are drugs in the WHO essential drugs list that have not been classified according to the BCS. This is because of the difficulty in obtaining permeability data, determined in vivo by absolute bioavailability or using in vitro, in silico and ex vivo models. Although for the registration of new drugs is necessary to define the absolute bioavailability, this is not usually easily accessible. As regards the other applicable models difficulties of standardization of methodologies, reproducibility of results and inconsistency with in vivo data are observed (Souza, Freitas, Storpirtis, 2007).

There are some criticisms of the use of BCS that fail in taking into account the dynamic system that occurs in vivo. An example of this is non-steroidal anti-inflammatory drugs that are classified as II and show extensive absorption. The biopharmaceutics drug disposition classification system (BDDCS), an alternative to the BCS, was proposed by Benet and collaborators (2008). It classifies drugs using aqueous solubility and metabolism (for the classification of permeability). This system aims to explain the results of BD in vivo for drugs with high permeability and also extensive metabolism. The correlation between metabolism and permeability can be explained in the case of drugs BCS of class I and II (highly permeable) that are able to reach the metabolic enzymes in hepatocytes. On the other hand, there are also limitations to BDDCS, among which the need of a suitable methodology that is able to predict the effect of the formulation components on the intestinal transporters. An example of the discrepancy of the results is sotalol, which has low liver metabolism, absorption less than 90%, and is approved by the FDA as Class I (Benet et al., 2008; Chen, Yu, 2009).

The same occurs to solubility data, although available in greater number in the literature, were not always obtained from experiments performed in accordance to biowaiver requirements as the means and temperature appropriate (Table I) (Lindenberg, Kopp, Dressman, 2004).

Furthermore, the BCS must be selectively used and considering carefully regulatory risks versus the benefits for a possible biowaiver. In particular, for compounds that do not have clarity on permeability data from the literature, the potential risk of rejection of the permeability study submitted or a potential delay due to issues that may arise during the review of the data, must be weighed against time and costs required to conduct a bioequivalence study (Cook, Addicks, Wu, 2008). It should also be demonstrated that the excipients included in the new formulation are well established for use in products containing such drug. These pharmaceutical adjuvants should not cause differences between the reference product and the generic candidate product in in the terms of affecting the absorption process, or other pharmacokinetic processes (WHO, 2006).

Noteworthy, the demonstration of bioequivalence is the biggest concern for the approval and use of generic products, and the possibility of a biowaiver for in vivo bioequivalence studies must be approached with caution and careful supervision to ensure the safety and efficacy of these drugs (Ramirez et al., 2010).

In practice, a universalization of the benefits of biowaiver based on BCS is not expected due to the existence of differences in regulations worldwide. For example, while the United States (FDA, 2000), the European Union (EMEA, 2008), Brazil (Brasil, 2011) and WHO (2006) allow biowaiver based on dissolution and classification by BCS, Japan does not allow it (Yamashita, Tachiki, 2008). At this point, the FDA's and EMA´s guide and the proposed requirements by WHO for biowaiver also show discrepancies in the definitions of high solubility (pH of 7.5 in the FDA's guide and 6.8 in EMA's guide and WHO proposal) and high permeability (the criterion of 90% on FDA's guide and 85% in the WHO proposal and EMA's guide) (Ramirez et al., 2010). Consequently, this leads to global pharmaceutical companies choosing to demonstrate the bioequivalence using a methodology that will enable acceptance by most countries. In general, there is a historical reluctance of these companies in continuing with biowaiver based on BCS due to uncertainty as to the success in their petitions to the various regulatory agencies (Cook et al., 2010).

The new drugs biowaiver employing the criterion of the BCS, according to Brazilian and U.S. regulations is indicated for solid oral dosage forms of immediate release, whose drugs are Class I (Brasil, 2011). It is also desirable that they are stable in the gastrointestinal tract, have wide therapeutic range, are not absorbed in the oral cavity, present low risk, their formulations contain excipients that do not significantly affect the rate and extent of oral absorption. Drugs are considered low-risk are the ones for which there are no reports of bioavailability or bioequivalence problems (United States, 2000).

According to the guidelines of WHO and EMA, Class III drugs can be included in the list of candidates for biowaiver. For this class, permeability is the factor that controls the absorption, so care should be taken since the rate and extent of absorption can be highly variable. The dissolution profile of the formulation should be clearly defined and reproducible (EMEA, 2008; WHO, 2006; Tsume, Amidon, 2010; Reddy, Karunakar, 2011).

For WHO, class II drugs are also likely biowaivers if they are weak acids showing high solubility at pH 6.8 and present, rapid or very rapid dissolving. For this class the limiting step of absorption is dissolution in vivo, thus the dissolution profile should be clearly defined and reproducible, emphasizing the importance of using methods that reflect or control the dissolution process in vivo (WHO, 2006; Alvarez et al., 2011).

Cristofoletti and colleagues (2013) calculated the diagnostic indicators utilizing 22 results of in vitro dissolution profiles performed at neutral conditions (phosphate buffer under pH 6.8-7.5) for a set of eight non-BE (all of them because of Cmax) and 14 BE drug products containing five weak acidic drugs, with pKa of less than 5.5 and which have dose numbers lower than one at pH values ranging from 6.8 to 7.4. However, these diagnostic parameters not were different from those calculated for all drugs belonging to BCS class II, in accordance with the previous reports for drug products containing ibuprofen that showed that dissolution tests, under neutral conditions, were unable to detect differences in absorption rate.

To minimize the risk of an incorrect decision about biowaiver in terms of public health and of individual risks to patients, the therapeutic indications of the drug, its pharmacokinetic variations, interactions with food, and other factors, should be evaluated (Mehta, 2007).

Exceptionally, excipients can have an impact on in vivo permeability, either directly in the active or passive passage of the drug through the gut wall or indirectly by altering the gastrointestinal transit time/residence time. The possible appearance of these effects is a risk that should not be ignored, but are relatively rare, and tend to require many specific high risk excipients to cause significant change in vivo (Butler, Dressman, 2010). It should be emphasized that a description of excipients is required, with a justification if the amount of each excipient is within the range considered normal. The so called active excipients such as, for example, sorbitol, mannitol, sodium lauryl sulfate or other surfactant must be identified when present in the medicine as well as their possible impact with respect to: gastrointestinal motility, sensitivity interaction with the drug, the drug permeability and interaction with membrane transporters. In those cases where critical excipients are relevant, the same amount should be used in both reference and generic products (EMEA, 2008).

CONCLUSION

One of the current applications of BCS is to supply means to provide biowaiver of new generic medicines containing drugs belonging to Class I (FDA, EMA, WHO and ANVISA), class III (EMA and WHO) and a specific group of compounds of class II (WHO). Granting of biowaiver based on criteria such as BCS eliminates unnecessary exposure of healthy individuals to drugs, reduces the regulatory burden, and provides economic relief, maintaining the high standard of public health for therapeutic equivalence (Cook et al., 2010). Especially in developing countries, where there are often insufficient resources to achieve the bioequivalence studies, the use of BCS becomes an important tool to ensure the efficiency and quality of pharmaceutical products (Benet et al., 2008).

In this article, it was found that the BCS, greatly expands the possibilities of biowaiver, contributing to give greater flexibility to the registration of generic medicines of assured efficacy and safety, thus providing increased supply of affordable treatment options, which meets the public policy expanding access to medicines to the population. Since the politic of generics was deployed in Brazil, in 1999, a huge amount of investment has been made in order to inform people that the generic products pass by bioequivalence test and they are as effectives and safe as the reference drug. In 2014, it was proposed that similar drugs are interchangeable with the reference drugs. According to the proposal, similar packaging hall include in its brand, 'equivalent product', symbolized by the acronym 'EQ'. The brand will enable consumers and physicians to identify products that have proof of equivalence and perform the same therapeutic function in relation to the reference (ANVISA, 2014). Therefore, it should be emphasized that a safe biowaiver decision should be made scientifically based on adequate and reliable experimental data, aiming not to put this politics into disrepute.

ACKNOWLEDGMENTS

This work was supported by PROPP/UFOP, Capes, FAPEMIG (APQ-00481_11 and Toxifar Network), and Anvisa (TC 007/2012).

REFERENCES

AGÊNCIA NACIONAL DE VIGILÂNCIA SANITÁRIA. ANVISA. Instrução Normativa - IN N0 2, de 14 de março de 2013. 'Dispõe sobre a lista de fármacos candidatos à bioisenção baseada no sistema de classificação biofarmacêutica (SCB) e dá outras providências'. Available at: <http://bvsms.saude.gov.br/bvs/saudelegis/anvisa/2013/int0002_14_03_2013.html>. [ Links ]

AGÊNCIA NACIONAL DE VIGILÂNCIA SANITÁRIA. ANVISA. Consulta Pública -N0 1, de 2014. 'Dispõe sobre medicamentos equivalentes'. Available at: <http://portal.anvisa.gov.br/wps/wcm/connect/20ddca8042979c349ae8db1bb0036de1/Conuslta+P%C3%BAblica+n%C2%B0++1+DIMON+.pdf?MOD=AJPERES>. Accessed on: Sept. 2013. [ Links ]

ALT, A.; POTTHAST, H.; MOESSINGER, J.; SICKMÜLLER, B.; OESER, H. Biopharmaceutical characterization of sotalol-containing oral immediate release drug products. Eur. J. Pharm. Biopharm., v.58, n.1, p.145-150, 2004. [ Links ]

ALVAREZ, C.; NUNEZ, I.; TORRADO, J. J.; GORDON, J.; POTTHAST, H.; GARCIA-ARIETA, A. Investigation on the possibility of biowaivers for ibuprofen. J. Pharm. Sci., v.100, n.6, p.2343-2349, 2011. [ Links ]

AMIDON, G. L.; LENNERNÄS, H.; SHAH, V. P.; CRISON, J R . A theoretical basis for a bio-pharmaceutical drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability.Pharm. Res., v.12, n.3, p.413-420, 1995. [ Links ]

ARAÚJO, L. U.; ALBUQUERQUE, K. T.; KATO, K. C.; SILVEIRA, G. S.; MACIEL, N. R.; SPÓSITO, P. A.; SILVA-BARCELLOS, N. M.; SOUZA, J.; BUENO, M.; STORPIRTIS, S. Medicamentos genéricos no Brasil: panorama histórico e legislação. Rev. Panam. Salud Publica, v.28, n.6, p.480-492, 2010. [ Links ]

BALIMANE, P. V.; CHONG, S.; MORRISON, R A . Current methodologies used for evaluation of intestinal permeability and absorption. J. Pharmacol. Toxicol. Methods, v.44, p.301-312, 2000. [ Links ]

BENET, L; AMIDON, G .L.; BARENDS, D.; LENNERNÄS, H.; POLLI, J.; SHAH, V.; STAVCHANSKY, S.; YU, L. The use of BDDCS in classifying the permeability of marketed drugs. Pharm. Res., v.52, p.483-488, 2008. [ Links ]

BRASIL. Resolução RDC nº135 de 29 de maio de 2003a. 'Regulamento técnico para medicamentos genéricos'. Available at: <http://www.anvisa.gov.br/legis/resol/2003/rdc/135_03rdc.htm>. Accessed on: 15 May, 2012. [ Links ]

BRASIL. Resolução RDC nº 134 de 29 de maio de 2003b. 'Dispõe sobre a adequação dos medicamentos já registrados'. Available at: <http://www.anvisa.gov.br/legis/resol/2003/rdc/134_03rdc.htm>. Accessed on: 15 May, 2012. [ Links ]

BRASIL. Objetivos de Desenvolvimento do Milênio. 2007. Available at: <http://www.ipea.gov.br/sites/000/2/download/TerceiroRelatorioNacionalODM.pdf>. Accessed on: 12 Jun. 2012. [ Links ]

BRASIL. Resolução RDC nº 31 de 11 de agosto de 2010. 'Dispõe sobre a realização dos Estudos de Equivalência Farmacêutica e de Perfil de Dissolução Comparativo.' Available at: <http://bvsms.saude.gov.br/bvs/saudelegis/anvisa/2010/res0031_11_08_2010.html>. Accessed on: 15 Mai. 2012. [ Links ]

BRASIL. Resolução RDC nº 37 de 3 de agosto de 2011. 'Guia para isenção e substituição de estudos de biodisponibilidade relativa/bioequivalência' Available at: <http://bvsms.saude.gov.br/bvs/saudelegis/anvisa/2011/res0037_03_08_2011.html>. Accessed on: 15 Mai. 2012. [ Links ]

BUTLER, J. M.; DRESSMAN, J B . The develop ability classification system: Application of biopharmaceutics concepts to formulation development. J. Pharm. Sci., v.99, n.12, p.4940-4954, 2010. [ Links ]

CHEN, M. L; YU, L. The use of drug metabolism for prediction of intestinal permeability. Mol. Pharm., v.6, n.1, p.74-81, 2009. [ Links ]

COOK, J. A.; ADDICKS, W.; WU, Y H . Application of the biopharmaceutical classification system in clinical drug development - an industrial view. AAPS , v.10, n.2, p.306-310, 2008. [ Links ]

COOK, J. A.; DAVIT, B. M.; POLLI, J E . Impact of Biopharmaceutics Classification System-Based Biowaivers. Mol. Pharm., v.7, n.5, p.1539-1544, 2010. [ Links ]

CRISTOFOLETTI, R.; CHIANN, C.; DRESSMAN, J.; STORPIRTIS, S. A Comparative Analysis of Biophar maceutics Classification system and biopharmaceutics drug disposition classfication system: a Cross-Sectional Survey with 500 bioequivalence studies. J. Pharm. Sci., v.102, n.9, p. 3136-3144, 2013. [ Links ]

DAHAN, A.; MILLER, J. M; AMIDON, G L . Prediction of Solubility and Permeability Class Membership: Provisional BCS Classification of the World's Top Oral Drugs. AAPS , v.11, n.4, p.740-746, 2009. [ Links ]

EMEA. European Agency for Evaluation of Medicinal Products, Committee for Proprietary Medicinal Products. Note for Guidance on the Investigation of Bioavailability and Bioequivalence. 2001. Available at: <http://www.ema.europa.eu/pdfs/human/qwp/140198enfin.pdf>. Accessed on: 23 Abr. 2011. [ Links ]

EMEA. European Medicines Agency, Committee for Medicinal Products for Human Use (CHMP). Guideline on the Investigation of Bioequivalence. 2008. Available at: <http://www.ema.europa.eu/pdfs/human/qwp/140198enrev1.pdf>. Accessed on: 30 Mai. 2012. [ Links ]

INTERNATIONAL PHARMACEUTICAL FEDERATION. FIP. Available at: <http://www.fip.org/bcs>. Accessed on: 4 Mai. 2012. [site]. [ Links ]

GRAFFNER, C. Regulatory aspects of drug dissolution from a European perspective. Eur J. Pharm. Sci., v.29, n.3-4, p.288-293, 2006. [ Links ]

KASIM, N. A.; WHITEHOUSE, M.; RAMACHANDRAN, C.; BERMEJO, M.; LENNERNÄS, H.; HUSSAIN, A. S.; JUNGINGER, H. E.; STAVCHANSKY, S. A.; MIDHA, K. K.; SHAH, V. P.; AMIDON, G. L Molecular properties of WHO essential drugs and provisional Biopharmaceutical Classification. Mol. Pharm., v.1, n.1, p.85-96, 2004. [ Links ]

LEACH, B.; PALLUZI, J. E.; MUNDERI, P. Prescription for healthy development: increasing access to medicines. London: United Nations Development Programme, 2005. Available at: <http://www.unmillenniumproject.org/reports/tf_essentialmedecines.htm>. Accessed on: 27 May 2012. [ Links ]

LENNERNÄS, H.; ABRAHAMSSON, B. The use of biopharmaceutic classification of drugs in drug discovery and development: current status and future extension. J. Pharm. Pharmacol., v.57, n.3, p.273-285, 2005. [ Links ]

LINDENBERG, M.; KOPP, S.; DRESSMAN, J B . Classification of orally administered drugs on the World Health Organization Model list of Essential Medicines according to the biopharmaceutics classification system. Eur. J. Pharm. Biopharm., v.58, n.2, p.265-278, 2004. [ Links ]

LÖBENBERG, R.; AMIDON, G L . Modern bioavailability, bioequivalence and biopharmaceutics classification system. New scientific approaches to international regulatory standards. Eur. J. Pharm. Biopharm., v.50, n.1, p.3-12, 2000. [ Links ]

MANADAS, R.; PINA, M. E.; VEIGA, F. A dissolução in vitro na previsão da absorção oral de fármacos em formas farmacêuticas de liberação modificada. Rev. Bras. Cienc. Farm., v.38, n.4, p.375-399, 2002. [ Links ]

MARTINEZ, M. N.; AMIDON, G L . A Mechanistic approach to understanding the factors affecting drug absorption: a review of fundamentals. J. Clin. Pharmacol., v.42, p.620-643, 2002. [ Links ]

MELO, M. G. M. A produção de medicamentos genéricos no Brasil: Dificuldades e perspectivas. Porto Alegre, 2005. 164p. [Dissertação de Mestrado. Faculdade de Farmácia. Universidade Federal do Rio Grande do Sul]. [ Links ]

MEHTA, M. FDA Regulatory use of BCS . In: AAPS workshop on BE, BCS, and beyond, 2007, North Bethesda, MD. p.21-23 Available at: http://www.aapspharmaceutica.com/meetings/files/90/04Mehta.pdf. Accessed on: may. 2011. . [ Links ]

MIRANDA, E. S.; PINTO, C. B. S.; REIS, A. L. A.; EMMERICK, I. C. M.; CAMPOS, M. R.; LUIZA, V. L.; OSORIO-DE-CASTRO, C G S. . Disponibilidade no setor público e preços no setor privado: um perfil de medicamentos genéricos em diferentes regiões do Brasil. Cad. Saúde Pública, v.25, n.10, p.2147-2158, 2009. [ Links ]

PANCHAGNULA, R.; THOMAS, N S . Biopharmaceutics and pharmacokinetics in drug research. Int. J. Pharm., v.201, n.2, p.131-150, 2000. [ Links ]

POLLI, J. E.; ABRAHAMSSON, B. S. I, YU, L. X.; AMIDON, G. L.; BALDONI, J. M.; COOK, J. A.; FACKLER, P.; HARTAUER, K.; JOHNSTON, G.; KRILL, S. L.; LIPPER, R. A.; MALICK, W. A.; SHAH, V. P.; SUN, D.; WINKLE, H. N.; WU, Y.; ZHANG, H. Summary Workshop Report: Bioequivalence, Biopharmaceutics Classification System, and Beyond. AAPS, v.10, n.2, p.373-379, 2008. [ Links ]

RAMIREZ, E.; LAOSA, O.; GUERRA, P.; DUQUE, B.; MOSQUERA, B.; BOROBIA, A. M.; LEI, S. H.; CARCAS, A. J.; FRIAS, J. Acceptability and characteristics of 124 human bioequivalence studies with active substances classified according to the Biopharmaceutic Classification System. Br. J. Clin. Pharmacol., v.70, n.5, p.694-702, 2010. [ Links ]

REDDY, B. B. K.; KARUNAKAR, A. Biopharmaceutics Classification System: a regulatory approach. Dissol. Technol., v.2, p.31-37, 2011. [ Links ]

RODRIGUES, P. O.; STULZER, H. K.; CRUZ, A. P.; FOPPA, T.; CARDOSO, T. M.; SILVA, M A S. . Equivalência Farmacêutica entre Comprimidos de propranolol Comercializados no Mercado Nacional. Infarma, v.18, n.3-4, p.16-21, 2006. [ Links ]

SOUZA, J.; FREITAS, Z. M. F.; STORPIRTIS, S. Modelos in vitro para determinação da absorção de fármacos e previsão da relação dissolução/absorção. Rev. Bras. Cienc. Farm., v.43, n.2, p.1-13, 2007. [ Links ]

STORPIRTIS, S.; CONSIGLIERI, V O . Biodisponibilidade e bioequivalência de medicamentos: aspectos fundamentais para o planejamento e execução de estudos. ver. Farm. Bioquim. Univ. São Paulo, v.31, n.2, p.63-70, 1995. [ Links ]

STORPIRTIS, S.; OLIVEIRA, P. G.; RODRIGUES, D.; MARANHÃO, D. Considerações biofarmacotécnicas relevantes na fabricação de medicamentos genéricos: fatores que afetam a dissolução e a absorção de fármacos. Rev. Bras. Cienc. Farm., v.35, n.1, p.1-16, 1999. [ Links ]

STORPIRTIS, S.; MARCOLONGO, R.; GASPAROTTO, F.S.; VILANOVA, C.M. A equivalência farmacêutica no contexto da intercambialidade entre medicamentos genéricos e de referência: bases técnicas e científicas. Brasília: ANVISA, 2004 Available at: <http://www.anvisa.gov.br/divulga/artigos/index.htm>. Accessed on: 01 August 2011. . [ Links ]

STORPIRTIS, S.; GAI, M. N. Biofarmacotécnica: princípios de biodisponibilidade, bioequivalência, equivalência farmacêutica, equivalência terapêutica e Intercambialidade de medicamentos . In: STORPIRTIS, S.; GONÇALEZ, J. E.; CHIANN, C.; GAI, M N. . (Eds.). Biofarmacotécnica. 1.ed. Rio de Janeiro: Guanabara Koogan, 2009. p.3-11. [ Links ]

TAKAGI, T.; RAMACHANDRAN, C.; BERMEJO, M.; YAMASHITA, S.; YU, L. X.; AMIDON, G L . A Provisional Biopharmaceutical Classification of the Top 200 Oral Drug Products in the United States, Great Britain, Spain, and Japan. Mol. Pharm., v.3, n.6, p.631-643, 2006. [ Links ]

TSUME., Y AMIDON., G L . The biowaiver extension for BCS class III drugs: The effect of dissolution rate on the bioequivalence of BCS class III immediate-release drugs predicted by computer simulation. Mol. Pharm., v.7, n.4, p.1235-1243, 2010. [ Links ]

UNITED STATES, U. S. Department of Health and Human Services, Food and Drug Administration. Center of Drug Evaluation and Research (CDER). Guidance for Industry. Dissolution Testing of Intermediate Release Solid Oral Dosage Forms, Rockville: FDA, 1997 Available at: <http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070237.pdf>. Accessed on: 25 May 2012. . [ Links ]

UNITED STATES, U. S Department of Health and Human Services, Food and Drug Administration. Center of Drug Evaluation and Research (CDER). Guidance for Industry Waiver of In Vivo Bioavailability and Bioequivalence Studies for Immediate-Release Solid Oral Dosage Forms Based on a Biopharmaceutics Classification System. Rockville: FDA, 2000 Available at: <http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm070246.pdf>. Accessed on: 25 May 2012. . [ Links ]

UNITED STATES, U. S. Department of Health and Human Services, Food and Drug Administration. Statement of Andrew C. von Eschenbach, M.D. Commissioner of the Food and Drug Administration 2007. Available at: <http://www.fda.gov/NewsEvents/Testimony/ucm154237.htm>. Accessed on: 02 Jun. 2012. [ Links ]

YAMASHITA, S.; TACHIKI, H. Analysis of risk factors in human bioequivalence study that incur bioinequivalence of oral drug products. Mol. Pharm., v.6, n.1, 48-59, 2008. [ Links ]

Bcs Classification Guidance

YU, L. X.; AMIDON, G. L.; POLLI, J. E.; ZHAO, H.; METHA, M. U.; CONNER, D. P.; SHAH, V. P.; LESKO, L. J.; CHEN, M. L.; LEE, V. H.; HUSSAIN, A S . Biopharmaceutics Classification System: The scientific basis for biowaiver extensions. Pharm. Res., v.19, n.7, p.921-925, 2002. [ Links ]

WORLD HEALTH ORGANIZATION. WHO. The World Medicines Situation 2004. Available at: <http://apps.who.int/medicinedocs/en/d/Js6160e/>. Accessed on: 27 May 2012. [ Links ]

WORLD HEALTH ORGANIZATION. WHO. Proposal to waive in vivo bioequivalence requirements for WHO Model List of Essential Medicines immediate-release, solid oral dosage forms. Technical Report Series, No 937, 40th Report, Annex 8 of WHO Expert Committee on Specifications for Pharmaceutical Preparations. 2006. Available at: <http://whqlibdoc.who.int/trs/WHO_TRS_937_eng.pdf>. Accessed on: 5 March 2012. [ Links ]

Received: September 12, 2013; Accepted: September 02, 2014

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.