Biopharmaceutics Classification System (BCS) and Biowaivers: Role in Drug Product Design
Anshu Sharma*1, CP Jain1 , MS Ashawat2
1Department of Pharmaceutical Sciences, M.L.S. University Udaipur(Rajasthan)
2B.N. Girls College of Pharmacy, Udaipur, (Rajasthan)
* Corresponding Author E-mail: anshukiransharma@yahoo.com
ABSTRACT
The introduction of the Biopharmaceutics Classification System (BCS) into the Food and Drug Administration is a major step forward to classify the biopharmaceutical properties of drugs and drug products. Based on the mechanistic approaches to the drug absorption and dissolution process, the BCS enables the regulatory bodies to simplify and improve the drug approval process. The knowledge of BCS of drug in a formulation can also be utilized by the formulation scientist to develop a more optimized dosage form.
KEY WORDS Biopharmaceutics Classification System, Biowaivers, IVIVC, solubility.
INTRODUCTION:
The Biopharmaceutics Classification System1 (BCS) is not only a useful tool for obtaining waivers for in vivo bioequivalence studies but also for decision making in
the discovery and early development of new drugs. It is
because BCS is based on a scientific framework describing the three rate limiting steps in oral absorption. The necessary steps for a drug to be absorbed are (a) release of drug from dosage forms,
(b) Maintenance of dissolved state throughout gastrointestinal (GI) track, and (c) Permeation of drug molecules through GI membrane into hepatic circulation. (d) Enterohepatic metabolism that influences the systemic availability as well as release of metabolites into the systemic circulation.
Y. Wu and L. Z. Benet2 proposed Biopharmaceutical Drug Disposition Classification System (BDDCS) completes the absorption process by including the fourth rate-limiting step of first pass effect. The determination of solubility, permeability, and metabolic stability have been fully integrated by most pharmaceutical companies as an integral part of high throughput screening (HTS) and lead optimization3. It is arguable that application of BCS in lead compound selection for optimal chemistry may be more important than using BCS in biostudy waiver at a later development stage. After all, the aim of pharmaceutical industry is to discover better compounds not in doing less biostudies. Biopharmaceutic Classification System, provides drug designer an opportunity to manipulate structure or physicochemical properties of lead candidates so as to achieve better "deliverability". Considering the facts for failure of New Chemical Entity (NCEs), drug research, once concentrating on optimizing the efficacy and safety of the leads, dramatically transformed in the past two decades
Goals of the BCS Guidance:
• To improve the efficiency of drug development and the review process by recommending a strategy for identifying expendable clinical bioequivalence tests.
• To recommend a class of immediate-release (IR) solid oral dosage forms for which bioequivalence may be assessed based on in vitro dissolution tests.
• To recommend methods for classification according to dosage form dissolution, along with the solubility and permeability characteristics of the drug substance
Purpose of the BCS Guidance:
• Expands the regulatory application of the BCS and recommends methods for classifying drugs.
If the regulatory utility of dissolution tests for IR products are to be expanded, their reliability must be improved4. For
IR products this may be achieved by considering the mechanistic relationships between drug dissolution, physico-chemical characteristics of drugs, gastrointestinal physiology and absorption or permeation processes. To this effect BCS provides, with minimal reliance on in vivo pharmacokinetic data, a rational mechanistic frame work for developing reliable dissolution tests for assessing bioequivalence.
• Explains when a waiver for in vivo bioavailability and bioequivalence studies may be requested based on the approach of BCS.
The BCS also provides a means for identifying when dissolution in vivo is likely or not likely to be rate- limiting and allows for managing risks associated with reliance on in vitro dissolution for bioequivalence assessment.
Biopharmaceutics Classification System (BCS)
This system, pioneered by Professor Amidon, all drug
molecules are classified into one of four classes based on their solubility and permeability through the intestinal cell layer. The classes are as follows:
Class1 : The drugs belongs to this class exhibit a high dissolution and absorption. The rate limiting step is drug dissolution and if dissolution is very rapid then gastric emptying rate becomes the from rate determining step5. Class I consists of water-soluble drugs that are well absorbed the gastrointestinal tract and, in general, have the preferred physicochemical properties. For immediate release dosage forms the absorption rate will be controlled by the gastric emptying rate. However, to secure constant high bioavailability, the dissolution rate must be relatively fast, or over 85% dissolution in 15 minutes.
Class 2: These drugs have a high absorption rate and a low dissolution therefore absorption is limited primarily by drug dissolution in the gastrointestinal tract. In vivo drug dissolution is then a rate limiting step for absorption except at a very high dose. Class II consists of water-insoluble drugs which, when dissolved, are well absorbed from the gastrointestinal tract. The dissolution rate in vivo is usually the rate- limiting step in drug absorption. Commonly drugs in this class have variable absorption due to the numerous formulation effects and in vivo variables that can affect the dissolution profile. Various formulation techniques are applied to compensate for the insolubility of the drugs and the consequent slow dissolution rate. These include formulation of the amorphous solid form, nanoparticles, surfactant addition, salt formation and
complexation.6 By such techniques the formulator tries to move the drugs from Class II to Class I without changing the intrinsic ability of the drug molecules to permeate biomembranes.
Class 3: These drugs have high dissolution and low absorption. In vivo permeability is rate limiting step for drug absorption. These drugs exhibit a high variation in the rate and extent of drug absorption. Since the dissolution is rapid, the variation is attributable to alteration of physiology and membrane permeability rather than the dosage form factors. Class III consists of water-soluble drugs that do not readily permeate biomembranes. For these drugs the Rate limiting factor in drug absorption is their permeability7. Including absorption enhancing excipients in their formulation can enhance their bioavailability.
Class 4: It consists of water-insoluble drugs which when solubilized do not readily penetrate biomembranes. These drugs are usually very difficult to formulate for effective oral delivery.
The interest in this classification system stems largely from its application in early drug development8 and then in the management of product change through its life-cycle. In early drug development knowledge of the class of a particular drug is an important factor influencing the decision to continue or stop its development. Obviously a low solubility/low permeability drug will never be presented as an orally administered product and will probably encounter serious formulation difficulties. A company wishing to produce an oral dosage form will wish to limit its development to those molecules that have high permeability. Increasingly, these considerations are incorporated from the very earliest phases, with the concept of property-based design being used in combinatorial chemistry to target production of compounds showing optimal properties. Subsequent in vitro models are then applied to evaluate the drug in early development.
In Vitro Dissolution Tests and Bioequivalence
Assessment: Need for BCS
The large regulatory interest in the BCS comes from its combination with in vitro – in vivo correlation (IVIVC)9 to manage the issues of product quality throughout its life- cycle. For example, orally administered class II drugs are expected to show a correlation between observed dissolution rate in vitro and rate and extent of absorption in vivo. The correlation is expected as it is the rate of release from the product that is the controlling factor in the absorptive process, the intestinal barrier not being rate-limiting. A reliable IVIVC can therefore be used to predict in vivo performance based on in vitro results. This tool would thus permit an assessment of the effect of change on in vivo product performance based on in vitro tests. This change is incurred for all products at several stages, for example: scale-up change of manufacturing site, excipients, suppliers etc. At its most extreme, this could be a change of manufacturer, i.e. a generic product, and raises the ultimate possibility of approval of certain generic drugs using comparative in vitro results only. For class I drugs, which dissolve rapidly and are highly permeable, bioavailability problems, are not to be expected and dissolution testing could suffice to assure bioequivalence in the circumstances discussed above.
On rare occasions an inverse in vitro - in vivo relationship, i.e., higher peak drug concentration in blood for a product that exhibits a relatively slow rate of dissolution in vitro, in a particular media, compared with another product, have also been observed. Such examples of "failure" of in vitro dissolution tests10 to signal bio-in-equivalence have hindered the use of dissolution tests for assessing bioequivalence between two pharmaceutically equivalent products. Comprehensive research studies designed to elucidate mechanistic reasons for such failures are generally not available in the public domain. Also, data from such failed studies are generally not submitted to the Agency. Possible causes for such differences may include; 1) inappropriate specification (dissolution test conditions, primarily media composition, and acceptance criteria), (2) presence of an excipient that may alter drug absorption, and (3) other reasons (for example, statistical type II error).
Experience gained through development of traditional in vitro - in vivo correlations (e.g., Level A, B, or C correlations) for immediate release (IR) products containing poorly soluble drugs and for extended release products suggests a significant degree of formulation dependency or specificity associated with such correlations. Therefore, for products that are likely to exhibit slow in vivo dissolution, in vitro - in vivo correlations need to be established and their predictive performance verified through experimentation. Future research in this area should address how to a priori identify dissolution test conditions that yield robust in vitro - in vivo correlations that are applicable to a wide range of formulations.
BIO-WAIVERS BASED ON THE BCS
The BCS are adopted by the World Health Organization (WHO), and classifies the drug molecules listed on the essential medicines list (EML)11 based on their solubility and permeability characteristics into four different classes. The use of in vitro testing to achieve a waiver of in vivo studies is commonly referred to as a biowaiver. Certain drug classes to be considered for a biowaiver, i.e. approval of products based on their in vitro drug dissolution tests instead of their human bioequivalence data, which is a costly task for drug manufacturers. By utilizing in vitro parameter such biowaivers significantly improve the speed and decrease the cost of bringing orally administered formulation to market. Currently, the BCS system allows a waiver of in vivo bioequivalence testing of immediate-release solid dosage forms for class 1 drugs. Whereas waivers for class 3 drugs are recommended only based on scientific justifications. Recent research has lead to the use of in-vitro tests to waive additional in vivo bioequivalency studies for some pharmaceutical products. The opportunities for cost-reduction through the limitation of costly in vivo studies make this a current and widely-debated topic. The BCS Working Group of the FDA proposed subsequent draft guidance that a ‘bio-waiver’ (i.e. assessment of bioequivalence using in vitro testing only) could be considered for class I drugs, provided they meet a list of criteria. Considering the uncertainties associated with in vitro dissolution tests, the proposed draft guidance recommends biowaivers only for rapidly dissolving products of highly soluble and highly permeable drugs that are not considered, by the FDA, to be "Narrow Therapeutic Index Drugs." The criterion for defining the therapeutic index of a drug is currently under consideration at the FDA. It is proposed that BCS based biowaivers apply for situations during both pre- (IND/NDA and ANDA) and post approval phases. The FDA guidance outlines five categories of biowaivers8.
1. Biowaivers without an IVIVC,
2. Biowaivers using an IVIVC: non-narrow therapeutic index drugs,
3. Biowaivers using an IVIVC: narrow therapeutic index drugs,
4. Biowaivers when in vitro dissolution is independent of dissolution test conditions and situations for which an IVIVC is not recommended for biowaivers
Biowaiver Consideration Active Pharmaceutical Ingredient (API)
In order to be considered bioequivalent according to the FDA Biowaiver procedure, a pharmaceutical product:
• Should contain a class 1 substance
• Should be rapidly dissolving, meaning it should release at least 85% of its content in 30 minutes in three different buffers (pH 1.2, pH 4.5 and pH 6.8) in a paddle (50 rpm) or basket (100 rpm) apparatus at 37°C and a volume of 900 ml
• Should not contain excipients, which could influence the absorption of the drug
• Should not contain a drug with a narrow therapeutic index
• Should not be designed to be absorbed from the oral cavity.
The reasoning for the above-mentioned class 1 substance, dissolution restriction is that when a highly soluble, highly permeable drug dissolves rapidly, it behaves like a solution in the gastrointestinal tract. If excipients give insignificant data, it should be excluded from the formulation because it influence the uptake across the gut wall and ultimately influence the bioavailability. The API is not prone to precipitation after its dissolution due to its good solubility under all pH conditions likely to be found in the upper gastrointestinal tract. The high drug permeability assures the complete uptake (> 90%) of the API during its passage through the small intestine. The fast dissolution of the product guarantees that the API is available long enough for the uptake in small intestine (the passage time in the small intestine is approximately 4 hours) and negates any slight differences between the formulations. Pharmaceutical products containing an API with a narrow therapeutic index should always be tested with in vivo methods, since the risk for the patient resulting from a possible incorrect bioequivalence decision using the Biowaiver procedure is considered too high with these kinds of APIs. As the BCS is only applicable to drugs which are absorbed from the small intestine, drugs with different sites of absorption (oral cavity) are not eligible for a Biowaiver. It can be easily seen that the FDA requirements for classification of APIs and eligibility criteria for the Biowaiver are very strict. In the last decade, several scientific data and continuing discussions have suggested that the original FDA criteria for application of the Biowaiver procedure can be relaxed without substantially increasing the risk to public health or to the individual patient. On the basis of these data and dialogue, the WHO has proposed revised BCS criteria and additional considerations for the eligibility of a pharmaceutical product for the Biowaiver procedure in the Multisource document12.
Protocol for Biowaivers
Required Data for Rapid and Similar Dissolution
• A brief description of the IR products used for dissolution testing.
• Dissolution data obtained with 12 individual units of the test and reference products at each specified testing interval for each individual dosage unit. A graphic representation of the mean dissolution profiles for the test and reference products in the three media (pH 1.2,4.5, 6.8).13
• Data supporting similarity in dissolution profiles between the test and reference products in each of the three media, using the f2 metric.
Required Data for High Permeable drugs:
• For human pharmacokinetic studies, information on study design and methods used along with the pharmacokinetic data.
• For direct permeability methods, information supporting method suitability with a description of the study method, criteria for selection of human subjects, animals, or epithelial cell line, drug concentrations, description of the analytical method, method to calculate extent of absorption or permeability, and information on efflux potential (if appropriate).
• A list of selected model drugs along with data on the extent of absorption in humans used to establish method suitability, permeability values and class for each model drug, and a plot of the extent of absorption as a function of permeability with identification of the low/high permeability class boundary and selected internal standard.
• Permeability data on the test drug substance, the internal standards, stability information, data supporting passive transport mechanism where appropriate, and methods used to establish high permeability of the test drug substance.
Required Data for Highly Soluble drugs:
• Information on chemical structure, molecular weight, nature of drug substance, dissociation constants.
• Description of test methods (analytical method, buffer composition).
• Test results summarized in a table with solution pH, drug solubility, and volume to dissolve highest dose strength.
• Graphical representation of mean pH-solubility profile
Methodology for the Dissolution Characteristics of a
Drug Product
The following approaches are recommended for classifying a drug substance and determining the dissolution characteristics of an IR drug product according to the BCS.
Class Boundaries
• A drug substance is considered HIGHLY SOLUBLE when the highest dose strength is soluble in < 250 ml water over a pH range of 1 to 7.5.
• A drug substance is considered HIGHLY PERMEABLE when the extent of absorption in humans is determined to be > 90% of an administered dose, based on mass-balance or in comparison to an intravenous reference dose.
• A drug product is considered to be RAPIDLY DISSOLVING when > 85% of the labeled amount of drug substance dissolves within 30 minutes using USP apparatus I or II in a volume of < 900 ml buffer solutions.
A. SOLUBILITY DETERMINATION
• pH - solubility profile of test drug in aqueous media with a pH range of 1 to 7.5.
• Shake-flask or titration method.
• Analysis by a validated stability-indicating assay.
An objective of the BCS approach is to determine the equilibrium solubility of a drug substance under physiological pH conditions. The pH-solubility profile of the test drug substance should be determined at 37 ± 1oC in aqueous media with a pH in the range of 1-7.5.14 A sufficient number of pH conditions should be evaluated to accurately define the pH-solubility profile. The number of pH conditions for a solubility determination can be based on the ionization characteristics of the test drug substance. Depending on study variability, additional replication may be necessary to provide a reliable estimate of solubility. Standard buffer solutions described in the USP are considered appropriate for use in solubility studies. Methods other than the traditional shake-flask method, such as acid or base titration methods, can also be used with justification to support the ability of such methods to predict equilibrium solubility of the test drug substance. Concentration of the drug substance in selected buffers (or pH conditions) should be determined using a validated stability-indicating assay that can distinguish the drug substance from its degradation products. The solubility class should be determined by calculating the volume of an aqueous medium sufficient to
Table 1: Suggested Model Drug to Classify Permeability of New Drug Candidate
Drug |
Permeability Class |
Comments |
Alpha-methyldopa Antipyrine Atenolol Caffeine Carbamazepine Hydrochlorothiazide Furosemide Ketoprofen Mannitol Metoprolol Naproxen Polyethylene glycol 400 Propanolol Ranitidine Theophylline Verapamil |
Low High Low High High Low Low High High to low High High Low High Low High High |
Amino acid transporter Permeability marker Paracellular, internal standard
Class IV Class IV Border marker High to low marker, internal standard
PEG 4000 can be used as a non-absorbable marker for in vivo studies Internal standard Candidate for characterization of P-glycoprotein efflux in in-vitro systems |
S.NO. BCS
CLASS Examples Human
Formulation Strategies Impact 1. Class 1 High
solubility
high permeability Metoprolol, Diltiazem, Verapamil, Propranolol,
Abacavir, Acetaminophen Simple capsule
or tablet No
major challenges for
immediate
release dosage
forms;
Controlled release
dosage forms
may be
needed
to limit rapid absorption profile 2. Class 2 Low solubility high permeability Phenytoin,
Danazol, Ketoconazole,
Mefenamic acid, Nifedinpine. Amiodarone,
Atorvastatin Micronized API
and surfactant;
Nano-particle technology;
Solid dispersion; Melt granulation/extrusion; Liquid or semisolid filled; capsule; Coating technology Formulations designed
to overcome dissolution
rate problems: Particle size reduction; Salt formation; Precipitation inhibitors; Metastable
forms;
Solid dispersion; Complexation; Lipid Technologies 3. Class 3 High
solubility
low permeability Cimetidine,
Acyclovir, Neomycin B,
Captopril. Ranitidine, Acyclovir, Atropine, ,
Cimetidine, Simple capsule
or tablet Absorption
enhancers Approaches
to improve
permeability: Prodrugs; Permeation Enhancers; Ion Pairing;
Bioadhesives Class 4 Low solubility low permeability Hydrochlorothiazide IV administrations Formulation would have
to use a combination of approaches
identified in Class
II and Class III
to overcome
dissolution and permeability problems Strategies
for oral administration
are not often
viable.
Use of alternative delivery
methods, such as intravenous
administration may
be most
effective
Table
2 BCS
Based Formulation Strategies:
Table 3: The Biopharmaceutics Classification System and Non-Oral Drug Delivery
S.NO. |
Drug Formulation |
D:S |
pH Range |
Dissolution Requirements |
1 |
Conventional oral tablets |
≤250m |
1 to 7.5 |
≥85% within 30 min |
2 |
Fast dissolving buccal tablets |
≤5 ml |
2 to 7.5 |
≈100% within 1 min |
3 |
Vaginal tablets |
≤5 ml |
3.5 to 4.9 |
- |
4 |
Pulmonary drug delivery |
≤0.5 ml |
6 to 7.6 |
Solution |
5 |
Aqueous nasal spray |
≤0.3 ml |
5 to 8.5 |
Solution |
6 |
Aqueous eye drop solution |
≤0.05 ml |
6.6 to 9 |
Solution |
dissolve the highest dose strength in the pH range of 1-7.5. A drug substance should be classified as highly soluble when the highest dose strength is soluble in <
250 ml of aqueous media over the pH range of 1-7.5. The reason for the 250 ml cut-off criterion for the dose: solubility ratio is that in pharmacokinetic bioequivalence studies, the API formulation is to be ingested with a large glass of water (8 ounces corresponds to about 250 ml)15. If the highest orally administered dose can be completely dissolved in this amount of water, independent of the physiological pH value (hence the determination over the pH range 1-7.5), solubility problems are not expected to hinder the uptake of the drug in the small intestine.
B. PERMEABILITY DETERMINATION
1. Extent of absorption in humans:
a) Mass-balance pharmacokinetic studies.
b) Absolute bioavailability studies.
2. Intestinal permeability methods:
• In vivo intestinal perfusions studies in humans.
• In vivo or in situ intestinal perfusion studies in animals.
• In vitro permeation experiments with excised human or animal intestinal tissue.
• In vitro permeation experiments across epithelial cell monolayers.
In many cases, a single method may be sufficient (e.g., when the absolute BA is 90% or more, or when 90% or more of the administered drug is recovered in urine). When a single method fails to conclusively demonstrate a permeability classification, two different methods may be advisable. Chemical structure and/or certain physicochemical attributes of a drug substance (e.g., partition coefficient in suitable systems) can provide useful information about its permeability characteristics. Sponsors may wish to consider use of such information to further support a classification.
Mass Balance Studies
Pharmacokinetic mass balance studies using unlabeled, stable isotopes or a radio labeled drug substance can be used to document the extent of absorption of a drug. Depending on the variability of the studies, a sufficient number of subjects should be enrolled to provide a reliable estimate of extent of absorption. Because this method can provide highly variable estimates of drug absorption for many drugs, other methods described below may be preferable.
Absolute Bioavailability Studies
Oral Bioavailability determination using intravenous administration as a reference can be used. Depending on the variability of the studies, a sufficient number of subjects should be enrolled in a study to provide a reliable estimate of the extent of absorption. When the absolute BA of a drug is shown to be 90% or more, additional data to document drug stability in the gastrointestinal fluid is not necessary.
Intestinal Permeability Methods
In vivo or in situ animal models and in vitro methods, such as those using cultured monolayers of animal or human epithelial cells, are considered appropriate for passively transported drugs. The observed low permeability of some drug substances in humans could be caused by efflux of drugs via membrane transporters such as P-glycoprotein (P-gp).16 When the efflux transporters are absent in these models, or their degree of expression is low compared to that in humans, there may be a greater likelihood of misclassification of permeability class for a drug subject to efflux compared to a drug transported passively. Expression of known transporters in selected study systems should be characterized. Functional expression of efflux systems (e.g., P-gp) can be demonstrated with techniques such as bidirectional transport studies, demonstrating a higher rate of transport in the basolateral-to-apical direction as compared to apical-to-basolateral direction using selected model drugs or chemicals at concentrations that do not saturate the efflux system (e.g., cyclosporin A, vinblastine, rhodamine 123). An acceptance criterion for intestinal efflux that should be present in a test system cannot be set at this time. Instead, this guidance recommends limiting the use of nonhuman permeability test methods for drug substances that are transported by passive mechanisms17.
To demonstrate suitability of a permeability method intended for application of the BCS, a rank-order relationship between test permeability values and the extent of drug absorption data in human subjects should be established using a sufficient number of model drugs. For in vivo intestinal perfusion studies in humans, six model drugs are recommended. For in vivo or in situ intestinal perfusion studies in animals and for in vitro cell culture methods, twenty model drugs are recommended. Depending on study variability, a sufficient number of subjects, animals, excised tissue samples, or cell monolayers should be used in a study to provide a reliable estimate of drug permeability. This relationship should allow precise differentiation between drug substances of low and high intestinal permeability attributes.
For demonstration of suitability of a method, model drugs should represent a range of low (e.g., < 50%), moderate (e.g., 50 - 89%), and high (≥ 90%) absorption. Sponsors may select compounds from the list of drugs and/or chemicals provided in (Table 1) or they may choose to select other drugs for which there is information available on mechanism of absorption and reliable estimates of the extent of drug absorption in humans.18
For a given test method with set conditions, selection of a high permeability internal standard with permeability in close proximity to the low/high permeability class boundary may facilitate classification of a test drug substance. For instance, a test drug substance may be determined to be highly permeable when its permeability value is equal to or greater than that of the selected internal standard with high permeability.
C. DISSOLUTION DETERMINATION
• USP apparatus I (basket) at 100 rpm or USP apparatus II (paddle) at 50 rpm.
• Dissolution media (900 ml): 0.1 N HCl or simulated gastric fluid, pH 4.5 buffer, and pH 6.8 buffer or simulated intestinal fluid.
• Compare dissolution profiles of test and reference products using a similarity factor (f2)
Dissolution testing should be carried out in USP Apparatus I at 100 rpm or Apparatus II at 50 rpm using 900 ml of the following dissolution media: (1) 0.1 N HCl or Simulated Gastric Fluid USP without enzymes; (2) a pH 4.5 buffer; and (3) a pH 6.8 buffer or Simulated Intestinal Fluid USP without enzymes. For capsules and tablets with gelatin coating, Simulated Gastric and Intestinal Fluids USP (with enzymes) can be used.
Dissolution testing apparatus used in this evaluation should conform to the requirements in USP . Selection of the dissolution testing apparatus (USP Apparatus I or II) during drug development should be based on a comparison of in vitro dissolution and in vivo pharmacokinetic data available for the product. The USP Apparatus I (basket method) is generally preferred for capsules and products that tend to float, and USP Apparatus II (paddle method) is generally preferred for tablets. For some tablet dosage forms, in vitro (but not in vivo) dissolution may be slow due to the manner in which the disintegrated product settles at the bottom of a dissolution vessel. In such situations, USP Apparatus I may be preferred over Apparatus II. If the testing conditions need to be modified to better reflect rapid in vivo dissolution (e.g., use of a different rotating speed), such modifications can be justified by comparing in vitro dissolution with in vivo absorption data (e.g., a relative BA study using a simple aqueous solution as the reference product).
A minimum of 12 dosage units of a drug product should be evaluated to support a biowaiver request. Samples should be collected at a sufficient number of intervals to characterize the dissolution profile of the drug product (e.g., 10, 15, 20, and 30 minutes). When comparing the test and reference products, dissolution profiles should be compared using a similarity factor (f ). The similarity factor5 is a logarithmic reciprocal square root transformation of the sum of squared error and is a measurement of the similarity in the percent (%) of dissolution between the two curves.
BCS Based Formulation Strategies
The BCS classification also used in the design19 of animal and human formulations. Animal formulation development is often more challenging than human formulation development due to the higher dose range required to explore toxicity for the establishment of adequate animal safety margins prior to human studies. It is not uncommon to dose animals up to a high dose of 1,000 or 2,000 mg/kg body weight whereas human doses are rarely as high as 1,000 mg per person or for a 70 kg person at 14 mg/kg body weight. The hurdle to solubilize the high animal doses often requires the use of vehicles containing organic solvents that can elicit toxicity by themselves. Human formulation has different requirements than animal formulation. A solid formulation (capsule/tablet) rather than a bottle of solution/suspension is a must for patient acceptability except in certain life-saving conditions. The amount of excipient used in a solid oral formulation is very limited (less than 1 g). Therefore excipient toxicity is rarely seen in human. The choice of excipients for human formulation is wider than those for animal formulation. However, the low weight of a tablet or the small volume of a capsule requires very high drug loadings. The solubilized formulation is often not feasible for high dose/low potency compounds. A comparison between animal and human formulation strategies is presented in Table 2.
The Biopharmaceutics Classification System and Non- Oral Drug Delivery
Although the BCS was originally developed for solid oral dosage forms a similar system can be applied to other types of drug formulations (Table 3). According to this system the solubility and dissolution requirements for many dosage forms are quite narrow when compared to the FDA’s requirements for immediate release solid dosage forms such as conventional tablets. A drug that is in Class I when given as an oral tablet can be in Class II when given in the form of nasal spray. For example, the solubility of diazepam in water (at room temperature) is 0.05 mg/ml. Diazepam and other benzodiazepines are primarily used as sedative-antianxiety drugs but they have also antiepileptic properties. Normal dose of diazepam is 5 mg. The D:S ratio is 100, which is well below the upper limit of 250 for oral delivery. The dose: solubility ratio15 indicates whether the capacity of the
f = 50 • log {[1 + (1/n)Σ
n 2 -0.5
(R - T ) ]
• 100}
GI fluids is sufficient to dissolve the entire dose
2 t=1 t t
Two dissolution profiles are considered similar when
administered. A dose: solubility ratio of 250 ml indicates
that conditions in the GI tract that sink conditions do not prevail. The oral bioavailability of diazepam has been
the f
2
value is 50. To allow the use of mean data, the
reported to be close to 100%. when given orally diazepam is
a Class I drug. However, if diazepam is to be formulated as
coefficient of variation should not be more than 20% at
the earlier time points (e.g., 10 minutes), and should not be more than 10% at other time points. Note that when both test and reference products dissolve 85% or more of the label amount of the drug in 15 minutes using all three dissolution media recommended above, the profile comparison with an f test is unnecessary.
2
nasal spray for treatment of seizures the upper limit of the D:S ratio is 0.3 ml, which makes it a Class II drug. Consequently formulation of aqueous diazepam nasal spray, possessing good bioavailability, will be quite difficult 20. Another example is 17β-estradiol. This drug has been given as 0.3 mg dose once a day (e.g., as vaginal cream) and its aqueous solubility (at room temperature) is 0.01 mg/ml. This gives D:S ratio of 30, which makes it a Class I drug when given orally. 17β-estradiol is a Class II drug when given by
other routes. Formulation of the drug as, for example, aqueous nasal spray (D:S ≤ 0.3) or as fast dissolving buccal tablet (D:S ≤ 5) is only possible through some
solubilizing techniques. Most new chemical entities
(NCE) are water-insoluble lipophilic compounds or, in other words, Class II or even Class IV compounds. It can be quite challenging for formulation scientists to create usable pharmaceutical products of such compounds.
CONCLUSION:
Regulatory application of BCS aids in the development of meaningful dissolution test specifications and for identifying immediate release solid oral products for which in vivo bioequivalence tests may not always be necessary. Regulation of IR products will be based on their intestinal permeability and solubility / dissolution characteristics. Similarly, the classification process can be envisioned to play an important role for regulating controlled release products, if adequate IVIVC is demonstrated. However, more research is still needed, for providing further support for these novel treatment concepts as well as advanced data analysis methods in order to continue to increase our understanding of pharmaceutical drug product performance in humans and to facilitate the drug regulation process based in this understanding. Bioavailability (BA) and bioequivalence (BE) play a central role in pharmaceutical product development and BE studies are presently being conducted for New Drug Applications (NDAs) of new compounds, The future revision of the BCS guidelines by the regulatory agencies in communication with academic and industrial scientists is exciting and will hopefully result in an increased applicability in drug development. Finally, we emphasize the great use of the BCS as a simple tool in early drug development to determine the rate-limiting step in the oral absorption process, which has facilitated the information between different experts involved in the overall drug development process. This increased awareness of a proper biopharmaceutical characterization of new drugs may in the future result in drug molecules with a sufficiently high permeability, solubility and dissolution rate, and that will automatically increase the importance of the BCS as a regulatory tool over time.
REFERENCES:
1. Amidon GL, et al. A theoretical basis for a biopharmaceutics drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm. Res. 1995; 12: 413–420.
2. ChiYuan W and 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.
3. Kerns EH, et al. Multivariate pharmaceutical profiling for drug discovery. Curr. Top. Med. Chem. 2002; 2:87–98.
4. Guidance for Industry, Dissolution Testing of Immediate Release Solid Oral Dosage Forms, FDA, CDER, August 1997, CDER/FDA.
5. Waterbeemd H and Lennernas H. Drug bioavailability: estimation of solubility, permeability, absorption and bioavailability, Wiley-VCH, Weinheim, Germany, 2003.
6. Luner C and Dressman JB. Improving drug solubility for oral delivery using solid dispersion, European Journal of Pharmaceutics and Biopharmaceutics. 2000; 50: 47-60.
7. Blume HH and Schug BS. The biopharmaceutics classification system (BCS):Class III drugs - better candidates for BA/BE waiver?, European J. of Pharma. Sci.1999; 9: 117–121
8. Sherry Ku M. Use of the biopharmaceutics classification system in early drug development. AAPS Journal. 2008; 10: 208.
9. Food and Drug Administration. Guidance for industry, waiver of in vivo bioavailability and bioequivalence studies for immediate-release solid oral dosage forms based on a biopharmaceutics classification system, Food and Drug Administration, Rockville, MD, 2000.
10. Dokoumetzidis A and Macheras P. A century of dissolution research: From Noyes and Whitney to the Biopharmaceutics Classification System. Intern. J. of Pharma.2006; 321: 1–11
11. World Health Organization. Proposal to waive in vivo bioequivalence requirements for WHO model list of essential medicines immediate-release, solid oral dosage forms. World Health Organization, Technical Report Series no. 937 .2006.
12. Multisource (Generic) Pharmaceutical Products: Guidelines on Registration Requirements to Establish Interchangeability. Working document AS/04.093/Rev. 4; WHO 2005
13. Dressman JB and Fleisher D. Mixing-tank model for predicting dissolution rate control of oral absorption. J. Pharm. Sci. 1986; 75: 109–116
14. Dressman JB, et al. Classification of orally administered drugs on the World Health Organization Model list of Essential Medicines according to the biopharmaceutics classification system, European Journal of Pharmaceutics and Biopharmaceutics. 2004; 58: 265–278.
15. Yu, LX., Amidon GL., et al. Biopharmaceutics classification system: the scientific basis for biowaiver extensions. Pharm Res. 2002; 19; 921.
16. Balimane PV, Han Y and Chong S. Current industrial practices of assessing permeability and p-glycoprotein interaction. The AAPS Journal. 2006; 8: 5.
17. Artursson P. Epithelial transport of drugs in cell culture. I: a model for studying the passive diffusion of drugs over intestinal absorptive aco-2) cells. J. Pharm. Sci.1990; 79: 476-482.
18. Lobenberg R and. Amidon GL. Modern Bioavailability, bioequivalence and biopharmaceutics classification system. New scientific approaches to international regulatory standards. Europ. J. of Pharma. and Biopharma. 2000; 50: 3.
19. Yu LX. Pharmaceutical Quality by Design: Product and Process Development, Understanding, and Control. Pharmaceutical Research. 2008; 25: 4.
20. Loftsson T. Cyclodextrins and the Biopharmaceutics Classification System of Drugs. J.Inclusion Phenomena and Macrocyclic Chem. 2002; 44: 63–67.
Received on 26.06.2008 Modified on 22.07.2008
Accepted on 20.08.2008 © RJPT All right reserved
Research J. Pharm. and Tech. 1(3): July-Sept. 2008; Page 144-151