Dissolution Method Validation Protocol

  1. Dissolution Method Validation Usp

The USP dissolution procedure is a performance test applicable to many dosage forms. It is one test in a series of tests that constitute the dosage form's public specification (tests, procedures for the tests, acceptance criteria). To satisfy the performance test, USP provides the general test chapters,. These chapters provide information about conditions of the procedure. For dissolution, these include information about (1) medium, (2) apparatus/agitation rate, (3) study design, (4) assay, and (5) acceptance criteria. Overall the dissolution procedure yields data to allow an accept/reject decision relative to the acceptance criteria, which are frequently based on a regulatory decision.

What are the other integral parts of analytical method validation? System suitability and acceptance criteria are integral parts of analytical validation. During each method/procedure, it should be ensured that all analytical operations, electronics and equipment are working properly at the time of analysis.

Jump to MATERIALS AND METHODS - The effect of the pH of the dissolution medium was studied. Preparation and optimized dissolution test procedure. In simple terms, a dissolution method transfer protocol (“protocol”) is a description of a mutual understanding of two parties, developer or current user of an analytical method (“originator”) and the receiver (“recipient”) of the method as to how a dissolution test is to be conducted.

This chapter provides recommendations on how to develop and validate a dissolution procedure. The procedure should be appropriately discriminating, capable of distinguishing significant changes in a composition or manufacturing process that might be expected to affect in vivo performance. It is also possible for the procedure to show differences between batches when no significant difference is observed in vivo. This situation requires careful evaluation of whether the procedure is too sensitive or appropriately discriminating. Assessing the results from multiple batches that represent typical variability in composition and manufacturing parameters may assist in this evaluation.

It is sometimes valuable to intentionally vary manufacturing parameters, such as lubrication, blend time, compression force, or drying parameters, to further characterize the discriminatory power of the procedure. A properly designed test should result in data that are not highly variable and should not be associated with significant analytical solution stability problems. High variability in results can make it difficult to identify trends or effects of formulation changes.

Dissolution results may be considered highly variable if the relative standard deviation (RSD) is greater than 20% at time points of 10 minutes or less and greater than 10% RSD at later time points. However, most dissolution results exhibit less variability than this.

The source of the variability should be investigated when practical, and attempts should be made to reduce variability whenever possible. The two most likely causes are the formulation itself (e.g., drug substance, excipients, or manufacturing process) or artifacts associated with the test procedure (e.g., coning, tablets sticking to the vessel wall or basket screen). Visual observations are often helpful for understanding the source of the variability and whether the dissolution test itself is contributing to the variability. Any time the dosage contents do not disperse freely throughout the vessel in a uniform fashion, aberrant results can occur.

Depending on the problem, the usual remedies include changing the apparatus type, speed of agitation, or deaeration; consideration and/or examination of sinker type; and changing the composition of the medium. Modifications to the apparatus may also be useful, with proper justification and validation. Many causes of variability can be found in the formulation and manufacturing process. For example, poor content uniformity, process inconsistencies, a reaction taking place at different rates during dissolution, excipient interactions or interference, film coating, capsule shell aging, and hardening or softening of the dosage form on stability may be sources of variability and interferences. During routine testing of the product, variability outside the expected range should be investigated from analytical, formulation, and processing perspectives. Physical and chemical data for the drug substance and dosage unit need to be determined before selecting the dissolution medium.

Two key properties of the drug are the solubility and solution state stability of the drug as a function of the pH value. When selecting the composition of the medium, the influence of buffers, pH value, and surfactants on the solubility and stability of the drug need to be evaluated.

Key properties of the dosage unit that may affect dissolution include release mechanism (immediate, delayed, or modified) and disintegration rate as affected by hardness, friability, presence of solubility enhancers, and presence of other excipients. Purified water is often used as the dissolution medium, but is not ideal for several reasons. First, the quality of the water can vary depending on the source of the water, and the pH value of the water is not controlled.

Second, the pH value can vary from day to day and can also change during the run, depending on the active substance and excipients. Despite these limitations, water is inexpensive, readily available, easily disposed of, ecologically acceptable, and suitable for products with a release rate independent of the pH value of the medium. The dissolution characteristics of an oral formulation should be evaluated in the physiologic pH range of 1.2 to 6.8 (1.2 to 7.5 for modified-release formulations). During method development, it may be useful to measure the pH before and after a run to discover whether the pH changes during the test. Selection of the most appropriate conditions for routine testing is then based on discriminatory capability, ruggedness, stability of the analyte in the test medium, and relevance to in vivo performance, where possible. The significance of deaeration of the medium should be determined, because air bubbles can interfere with the test results, acting as a barrier to dissolution if present on the dosage unit or basket mesh. Further, bubbles can cause particles to cling to the apparatus and vessel walls.

On the other hand, bubbles on the dosage unit may increase buoyancy, leading to an increase in the dissolution rate, or may decrease the available surface area, leading to a decrease in the dissolution rate. A dearation method is described as a footnote in the Procedure section under. Typical steps include heating the medium, filtering, and drawing a vacuum for a short period of time. Other methods of deaeration are available and in routine use throughout the industry.

Dissolution

Media containing surfactants are not usually deaerated because the process results in excessive foaming. To determine whether deaeration of the medium is necessary, results from dissolution samples run in nondeaerated medium and deaerated medium should be compared. Biorelevant medium is a medium that has some relevance to the in vivo performance of the dosage unit. Choice of a biorelevant medium is based on (1) a mechanistic approach that considers the absorption site, if known, and (2) whether the rate-limiting step to absorption is the dissolution or permeability of the compound. In some cases, the biorelevant medium will be different from the test conditions chosen for the regulatory test, and the time points are also likely to be different. If the compound dissolves quickly in the stomach and is highly permeable, gastric emptying time may be the rate-limiting step to absorption.

In this case, the dissolution test should demonstrate that the drug is released quickly under typical gastric (acidic) conditions. On the other hand, if dissolution occurs primarily in the intestinal tract (e.g., for a poorly soluble, weak acid), a higher pH range (e.g., simulated intestinal fluid with a pH of 6.8) may be more appropriate.

The fed and fasted states may also have significant effects on the absorption or solubility of a compound. Compositions of media that simulate the fed and fasted states can be found in the literature. These media reflect changes in pH, bile concentrations, and osmolarity after meal intake and therefore have a composition different from that of typical compendial media. They are primarily used to establish in vitro–in vivo correlations during formulation development and to assess potential food effects and are not intended for quality control purposes. For quality control purposes, the substitution of natural surfactants (bile components) with appropriate synthetic surfactants is permitted and encouraged because of the expense of the natural substances and the labor-intensive preparation of the biorelevant media.

Dissolution Method Validation Usp

When Apparatus 1 or 2 is not appropriate, another official apparatus may be used. Apparatus 3 (Reciprocating Cylinder) has been found to be especially useful for bead-type modified-release dosage forms. Apparatus 4 (Flow-Through Cell) may offer advantages for modified-release dosage forms that contain active ingredients with limited solubility. In addition, Apparatus 3 or Apparatus 4 may have utility for soft gelatin capsules, bead products, suppositories, or poorly soluble drugs.

Apparatus 5 (Paddle over Disk) and Apparatus 6 (Rotating Cylinder) have been shown to be useful for evaluating and testing transdermal dosage forms. Apparatus 7 (Reciprocating Holder) has been shown to have application to nondisintegrating oral modified-release dosage forms, as well as to transdermal dosage forms. Some changes can be made to the apparatus; for example, a basket mesh size other than the typical 40-mesh basket (e.g., 10, 20, 80 mesh) may be used when the need is clearly documented by supporting data.

In countries where available mesh sizes vary from the USP-specified mesh value, basket material with the nearest metric dimension should be used. Care must be taken that baskets are uniform and meet the dimensional requirements specified under. If the basket screens become clogged during dissolution of capsule or tablet formulations, it may be advisable to switch to the paddle method.

The volume can be increased from the typical 900 to 1000 mL by using 2- and 4-L vessels to assist in meeting sink conditions for poorly soluble drugs. A noncompendial apparatus may have some utility with proper justification, qualification, and documentation of superiority over the standard equipment.

For example, a small-volume apparatus with mini paddles and baskets may be considered for low-dosage strength products. The rotating bottle or static tubes (jacketed stationary tubes enclosed with a water jacket and equipped with a magnetic stirrer) may also have utility for microspheres and implants, peak vessels for eliminating coning, and modified flow-through cells for special dosage forms, including powders and stents. When sinkers are used, a detailed description of the sinker must be stated in the written procedure. It may be useful to evaluate different sinkers, recognizing that sinkers can significantly influence the dissolution profile of a dosage unit. When transferring the procedure, the sinkers should be duplicated as closely as possible in the next facility. There are several types of commercially available sinkers. A method for making sinkers by hand, sinkers that are similar to “a few turns of wire helix” as described in Apparatus 2 (Paddle Apparatus) under, is described below.

Rates outside 25 to 150 rpm are usually inappropriate because of the inconsistency of hydrodynamics below 25 rpm and because of turbulence above 150 rpm. Agitation rates between 25 and 50 rpm are generally acceptable for suspensions. For dosage forms that exhibit coning (mounding) under the paddle at 50 rpm, the coning can be reduced by increasing the paddle speed to 75 rpm, thus reducing the artifact and improving the data. If justified, 100 rpm may be used, especially for extended-release products.

Decreasing or increasing the apparatus rotation speed may be justified if the profiles better reflect in vivo performance and/or the method results in better discrimination without adversely affecting method reproducibility. For immediate-release dosage forms, the duration of the procedure is typically 30 to 60 minutes; in most cases, a single time point specification is adequate for Pharmacopeial purposes.

Industrial and regulatory concepts of product comparability and performance may require additional time points, which may also be required for product registration or approval. A sufficient number of time points should be selected to adequately characterize the ascending and plateau phases of the dissolution curve. According to the Biopharmaceutics Classification System referred to in several FDA Guidances, highly soluble, highly permeable drugs formulated with rapidly dissolving products need not be subjected to a profile comparison if they can be shown to release 85% or more of the active drug substance within 15 minutes.

For these types of products a one-point test will suffice. However, most products do not fall into this category. Dissolution profiles of immediate-release products typically show a gradual increase reaching 85% to 100% at about 30 to 45 minutes. Thus, dissolution time points in the range of 15, 20, 30, 45, and 60 minutes are usual for most immediate-release products. For rapidly dissolving products, including suspensions, useful information may be obtained from earlier points, e.g., 5 to 10 minutes.

For slower-dissolving products, time points later than 60 minutes may be useful. Dissolution test times for compendial tests are usually established on the basis of an evaluation of the dissolution profile data. So-called infinity points can be useful during development studies. To obtain an infinity point, the paddle or basket speed is increased at the end of the run for a sustained period (typically 15 to 60 minutes), after which time an additional sample is taken. Although there is no requirement for 100% dissolution in the profile, the infinity point can provide data that may supplement content uniformity data and may provide useful information about formulation characteristics during initial development or about method bias. For an extended-release dosage form, at least three test time points are chosen to characterize the in vitro drug release profile for Pharmacopeial purposes.

Additional sampling times may be required for drug approval purposes. An early time point, usually 1 to 2 hours, is chosen to show that there is little probability of dose dumping. An intermediate time point is chosen to define the in vitro release profile of the dosage form, and a final time point is chosen to show the essentially complete release of the drug. Test times and specifications are usually established on the basis of an evaluation of drug release profile data. For products containing more than a single active ingredient, drug release is to be determined for each active ingredient. Visual observations and recordings of product dissolution and disintegration behavior are very useful because dissolution and disintegration patterns can be indicative of variables in the formulation or manufacturing process.

To accomplish visual observation, proper lighting (with appropriate consideration of photodegradation) of the vessel contents and clear visibility in the bath are essential. Documenting observations by drawing sketches and taking photographs or videos can be instructive and helpful for those who are not able to observe the real time dissolution test. Observations are especially useful during method development and formulation optimization. Examples of typical observations include, but are not limited to, the following.Uneven distribution of particles throughout the vessel.

This can occur when particles cling to the sides of the vessel, when there is coning or mounding directly under the apparatus, when particles float at the surface of the medium, when film-coated tablets stick to the vessel, and/or when off-center mounds are formed.Air bubbles on the inside of the vessel or on the apparatus or dosage unit. Sheen on the apparatus is also a sign of air bubbles. Filter validation may be accomplished by preparing a suitable standard solution or a completely dissolved sample solution (e.g., prepared as a typical sample in a vessel or a sample put in a beaker and stirred with a magnetic stirrer for 1 hour). For standard solutions, compare the results for filtered solutions (after discarding the appropriate volume) to those for the unfiltered solutions. For sample solutions, compare the results for filtered solutions (after discarding the appropriate volume) to those for centrifuged, unfiltered solutions. The usual assay for a dissolution sample is either spectrophotometric determination or HPLC.

The preferred method of analysis is spectrophotometric determination because results can be obtained faster, the analysis is simpler, and fewer solvents are used. HPLC methods are used when there is significant interference from excipients or among drugs in the formulation to improve analytical sensitivity and/or when the analysis can be automated. It may be useful to obtain data for the drug with a stability-indicating assay (e.g., HPLC chromatograms) in the medium of choice, even if the primary assay is based on a spectrophotometric method. The validation topics described in this section are typical but not all-inclusive. The validation elements addressed may vary, depending on the phase of development or the intended use for the data.

The acceptance criteria are presented as guidelines only and may differ for some products. Firms should document the appropriate acceptance criteria for their products in pertinent SOPs. Other considerations may be important for special dosage forms. The extent of validation depends on the phase of the product development. Full validation takes place by the time of Phase III clinical studies.

Validation studies should address the variations associated with different profile time points. For products containing more than a single active ingredient, the dissolution method needs to be validated for each active ingredient. If the placebo interference exceeds 2%, then method modification—such as (1) choosing another wavelength, (2) baseline subtraction using a longer wavelength, or (3) using HPLC—may be necessary in order to avoid the interference. When other active drugs or significant levels of degradates are present, it is necessary to demonstrate that these do not significantly affect the results. One procedure for doing this is to measure the matrix in the presence and absence of the other active drug or degradate: any interference should not exceed 2%.

In cases of poor drug solubility, it may be appropriate to prepare a stock solution by dissolving the drug substance in a small amount of organic solvent (typically not exceeding 5%) and diluting to the final concentration with dissolution medium. An amount of stock solution equivalent to the targeted label claim may be added to the vessel instead of the drug powder. Similarly, for very low strengths, it may be more appropriate to prepare a stock solution than to attempt to weigh very small amounts. The measured recovery is typically 95% to 105% of the amount added. Bracketing or matrixing of multiple strengths may be useful. Intermediate Precision—Intermediate precision may be evaluated to determine the effects of random events on the precision of the analytical procedure. This evaluation is typically done later in the development of the drug product.

The precision can be across the range of product strengths. Typical variations to study include days, analysts, and equipment. The use of an experimental matrix design is encouraged for evaluation of intermediate precision. If possible, intermediate precision can be evaluated using a well-characterized lot of drug product of tight content uniformity. In cases where a well-characterized product is not available, placebo and active ingredient may be used to identify intermediate precision. Samples may be automatically introduced into the spectrophotometer using autosippers and flow cells. Routine performance checks, cleaning, and maintenance as described in the standard operating procedures or metrology documents are useful for reliable operation of these instruments.

Dissolution method validation protocol template

Cells with path lengths ranging from 0.02 to 1 cm are typically used. Cell alignment and air bubbles could be sources of error.

The smaller path length cells are used to avoid diluting the sample; however, acceptable linearity and standard error need to be demonstrated. For HPLC analysis, the compatibility of dissolution media and mobile phase may be examined, especially if large injector volumes (over 100 µL) are needed.

Samples are normally analyzed with HPLC using a spectrophotometric detector and an auto-injector. Single injections of each vessel time point with standards throughout the run constitute a typical run design. System suitability tests include, at a minimum, the retention window and injection precision. Typically, the repeatability of an HPLC analysis should be less than or equal to 2% RSD for five or six standard determinations. The standard level is typically at the 100% label claim level, especially for a single-point analysis.

Preparation of the placebo samples for the HPLC analysis is to be performed in the same way as in the spectrophotometric analysis. Examine the chromatogram for peaks eluting at the same retention time as the drug.

If there are extraneous peaks, inject the standard solution, and compare retention times. If the retention times are too close, spike the placebo solution with the drug. Chromatograms may also be obtained over an extended run time using the blank (dissolution medium), standard, and sample solution to identify late eluters that may interfere with subsequent analyses. Typical acceptance criteria for the amount of active ingredient dissolved, expressed as a percentage of the labeled content (Q), are in the range of 75% to 80% dissolved. A Q value in excess of 80% is not generally used, because allowance needs to be made for assay and content uniformity ranges.

Acceptance criteria including test times are usually established on the basis of an evaluation of the dissolution profile data. Acceptance criteria should be consistent with historical data, and there is an expectation that acceptable batches (e.g., no significant differences in in vivo performance, composition, or manufacturing procedure) will have results that fall within the acceptance criteria.