Challenges of Dissolution Methods Development for Soft ...

08 Jul.,2024

 

Challenges of Dissolution Methods Development for Soft ...

Noyes and Whitney first documented the study of the dissolution process in as a field of physical chemistry, which later was mimicked in pharmacy due to its importance in drug administration [74]. The dissolution of solid dosage forms attracted attention as the realization of the importance of drug dissolution concerning bioavailability was identified in the s with the understanding that only dissolved drugs can diffuse through the human body [74,75,76,77,78]. Poor drug solubility and low dissolution rates potentially lead to insufficient availability of the drug at the site of action and subsequent failure of the in vivo therapeutic performance. This is independent of the fact that the drug could be an ideal structure for the target site. Essentially, if the drug is too insoluble, it can never reach its target site, and it will be of no therapeutic relevance. Characterization of the dissolution of a drug from a given dosage form is critical for the successful development of a drug product. This section discusses the current state-of-the-art of SGCs dissolution and various practical concepts of developing dissolution methods for SGCs.

If you want to learn more, please visit our website.

Dissolution testing is an official test used for evaluating the rate of drug release from a dosage form into the dissolution medium or solvent under standardized conditions of liquid/solid interface, temperature, paddle speed, or solvent composition. Dissolution testing has become important in measuring the in vitro rate and extent of API release from different dosage forms, including SGCs. Dissolution can be described as a process by which molecules of a solute (e.g., API) are dissolved in a solvent to form a solution. The in vivo effectiveness of a dosage form depends on its ability to release the drug for systemic absorption. SGCs dissolution goes through three main steps, the first one being swelling and rupture of the gelatin shell, followed by release and dispersion of the fill material, and finally, the dissolution of the active ingredient(s) in the dissolution medium ( ). These processes occur in series, and thus the slowest step determines dissolution rate of the SGCs. The slowest step in this case controls the overall rate and extent of drug absorption. However, this varies from drug to drug. For poorly soluble drugs, especially BCS II and IV, their dissolution will be a rate-limiting step in the absorption process. On the other hand, for drugs that have high solubility, their dissolution will be rapid, and rate and extent of absorption can be affected by other factors, e.g., membrane permeability, enzymes degradation in the GIT, or first pass metabolism.

A critical requirement for drug products is that they release the APIs in vivo at a predictable rate [ 9 , 82 , 83 ]. The kinetics of drug release follows the release mechanism of the system, such as diffusion through the inert matrix, diffusion across the gel, osmotic release, ion-exchange, or pH-sensitive delivery systems. Among the various mechanisms involved in API release, diffusion is the principal release mechanism, and it takes place at varying degrees in every system. Solute release models in physical chemistry preceded the development of drug delivery systems by many years [ 77 , 78 ]. In , Higuchi introduced a mathematical model of drug release for diffusion-controlled systems [ 84 ]. The author analyzed the release kinetics of an ointment, assuming that it is homogeneously dispersed and is released in the planar matrix and the medium. According to the model, the release mechanism is proportional to the square root of time [ 85 ]. This model is recommended for the initial 60% of the release curve due to its approximate nature. In late , Wang published an article considering the two independent mechanisms of transport, Fick&#;s law, and polymer relaxation on the molecules&#; movement in the matrix [ 86 ]. Then, Peppas, in , introduced a semi-empirical equation, power law, to describe drug release from polymeric devices in a generalized way [ 87 , 88 ].

Another concept that needs to be introduced here is the drug release phenomenon. Drug dissolution rates and drug release rates are quite different. Drug release refers to the process by which the drug in a drug product is released in the dissolution medium or at the site of absorption by diffusion or dissolution of a drug product. Depending on the physical form of the API in the drug product, the release of API may be slow or immediate. As described in the previous section, dissolution is a process by which molecules of a solute are dissolved in solvent vehicles as a function of time. On the other hand, the term &#;release&#; most often refers to a much more complex phenomenon. Release encompasses capsule dissolution as one of its several steps. Upon contact with the aqueous medium, water penetrates the soft gelatin shell and at least partially dissolves the API [ 81 ]. Then, the dissolved API diffuses out through the capsule shell due to concentration gradients. Furthermore, the gelatin shell might undergo significant swelling as soon as the critical water content is reached, which will result in the rupture of the shell, followed by dispersion and eventual dissolution in the release medium. Hence, several steps are involved in the process of releasing the API from SGCs drug products, with only one of them being drug dissolution.

The dissolution rate of a drug product in each solvent is defined as the rate of transfer of individual drug molecules from the solid particles into the solution as individual molecules, and it can be expressed as the concentration of dissolved API for a given time interval. The rate of dissolution can vary depending on the form of API, e.g., the amorphous form usually has rapid dissolution compared to crystalline forms of API [ 79 , 80 ].

Another important thermodynamic property in a discussion of dissolution processes is solubility, which may be expressed in several ways, including but not limited to molarity, molality, mole fraction, mole ratio, and parts per million. As an illustration, for the case of a drug molecule, consider an excess amount of solid that is exposed to the solvent phase at a defined temperature and pressure. In the equilibrium state, the number of drug molecules going into the solution equals the number of drug molecules which re-precipitate. Under these conditions, the solution is saturated with drug molecules and the concentration of dissolved drug under these conditions is defined as the &#;equilibrium drug solubility&#; (specific to the given temperature and pressure) [ 89 ]. It is important to assure that the solid phase present at the beginning of the experiment remains unaltered after reaching thermodynamic equilibrium during any solubility experiment. It is worth mentioning that, when particle size or the presence of additives, or the pH modifies the intrinsic solubility, this is usually reported as &#;apparent solubility&#; to distinguish it from the equilibrium value. In order to avoid the inconsistency in solubility data reporting, the size of filters used in the separation of dissolved drug particles must be stated.

However, the USP General Chapter <>, Disintegration and dissolution of dietary supplements, accepts a rupture test as a performance test of SGCs if the capsule content is semi-solid or liquid [ 92 ]. The rupture test is performed using apparatus 2, as described under General Chapter Dissolution <711>, at a rotation speed of 50 rpm in 500 mL of immersion medium for a duration of 15 min. As per USP <>, the requirements are met if all of the SGCs tested rupture in not more than 15 min&#;. If 1 or 2 of the SGCs rupture in more than 15 min but not more than 30 min, the test is repeated on 12 additional SGCs: not more than 2 of the total of 18 capsules tested rupture in more than 15 but not more than 30 min. For SGCs that do not conform to the above rupture test acceptance criteria, the test is repeated with the addition of papain to the medium in the amount that results in an activity of not more than 550,000 units/L of medium or with the addition of bromelain in the amount that results in an activity of not more than 30 gelatin-digesting units/L of medium [ 92 ]. Almukainzi et al. [ 93 ] compared the rupture and disintegration tests of SGCs of amantadine, ginseng, flaxseed oil, pseudoephedrine hydrochloride, and soybean oil. Their data showed that neither rupture test nor disintegration test was advantageous over the other. However, rupture test reached the endpoint quicker compared to the disintegration test. In another study, Bachour et al. [ 94 ] evaluated the suitability of the rupture test for stability studies of SGCs containing oil-based oral multivitamins. Their study showed that the rupture test was sensitive to stability conditions, and that the commercial drug products passed the rupture test. However, all long-term stability samples failed the rupture test using tier 2 conditions. This indicates that the rupture test may be suitable for assessing the performance of some drug products, but this will depend on the properties of fill components.

The disintegration test is considered as one of the performance tests for the immediate release dosage forms [ 90 ]. As per the USP <701>, disintegration is defined as &#;the state in which any residue of the unit, except fragments of insoluble coating or capsule shell, remaining on the screen of the test apparatus or adhering to the lower surface of the disk, if used, is a soft mass having no palpably firm core&#; [ 91 ]. The requirements of disintegration are met if all test units have completely disintegrated or if not fewer than 16 of a total of 18 units tested are disintegrated within a predetermined time period. This does not imply complete solution of the API or the drug product.

6.5. Practical Concepts of Developing a Dissolution Method

Dissolution testing is used throughout drug product development as an indicator of drug product performance. During formulation development, dissolution testing is used to demonstrate the release and uniformity of a dosage form in a simulated environment. Once the performance is established for the product, this information is used periodically during stability to determine if the characteristics of the product are changing in such a way that the product continues to or stops performing as required. Often, the performance of a drug product in dissolution shows physical behavior; however, it does not necessarily indicate performance in vivo. Therefore, correlation between dissolution and pharmacokinetic data can be used to demonstrate if dissolution testing has the ability to predict drug performance. This is referred to as establishing in vitro&#;in vivo correlation (IVIVC) [95].

The purpose of this section is to give an overview of the practical concepts of developing dissolution test methods for SGCs. It is important to understand that the dissolution of a product requires a number of physical changes to take place. Unlike other typical solid dose forms, SGCs must first reach the point where the integrity of the gelatin is compromised and the outer shell ruptures to allow release of the fill material. Following this, the fill components must disperse within the media to allow the active ingredients to either enter solution or distribute evenly throughout the media ( ). The challenge is that the capsule shell is very sensitive to its environment and can change relative to hardness, cross-linking, and seam integrity, which can all play a role in perceived dissolution changes when in fact they are changes in rupture time. Therefore, it is essential to develop a dissolution strategy that accounts for differences in the integrity of the capsule shell as well as changes in the fill material.

Dissolution methods development are labor-intensive processes even with careful technique and practice. It is important to invest time in developing a procedure that can be efficiently executed on a routine basis and repeated robustly. Dissolution tests are required by the Pharmacopeias to determine the release of the drug from the dosage form in an environment with a pH from 1.2 to 7.4. For example, USP <711> [96] requires a two-step dissolution method for enteric-coated solid oral dosage forms that demonstrates coating integrity in an acidic environment, usually 0.1 N HCl, followed by exposure to a neutral pH environment, preferably with a phosphate buffer, where the first step of dissolution method provides information about the coating quality and the potential for coating failure. The United States Pharmacopeia (USP) and the U.S. Food and Drug Administration (FDA) provide guidelines on the development and validation of dissolution procedures [96,97]. Most of these guidelines are for solid oral dosage forms like tablets and hard gelatin capsules; however, one cannot extrapolate these methods to SGCs without proper assessment. The choice of dissolution method should be based on the dosage form and the fill characteristics of SGCs. shows the common USP dissolution apparatus used in dissolution testing.

Table 2

Type of ApparatusPrincipleCommon Dosage FormsType 1BasketIR, chewable tablets, DR, ER, suppositories, capsules, floating dosage formsType 2PaddleIR, ODT, chewable tablets, DR, ER, enteric-coated tablets or capsulesType 3Reciprocating CylinderCR, chewable tablets and beadsType 4Flow-Through CellER, soft and hard gelatin capsules, powder, granules, pellets, suppositories, and implantsType 5Paddle Over DiskTransdermal patches, ointments, and emulsionsType 6Rotating CylinderTransdermal patchesType 7Reciprocating HolderTransdermal formulations, and ND oral-modified release formulationsOpen in a separate window

Developing a discriminating dissolution test for SGCs requires special considerations and knowledge of gelatin and fill material properties and factors influencing them. Several factors affect the dissolution behavior of SGCs and subsequently affect the development of dissolution procedures. These factors include physical properties of the gelatin shell, physical and chemical properties of the fill material, chemical interaction between the gelatin shell and fill components, and moisture exchange between the shell and the fill material. In particular, moisture exchange can potentially result in brittleness of the gelatin shell, and chemical interactions between the shell and fill could result in gelatin cross-linking.

Two key considerations in the design and development of dissolution methods are the solubility of the active ingredient and solution stability of the SGCs. To establish a suitable medium, several dissolution media should be evaluated to identify the one that achieves appropriate sink conditions. Sink conditions can be defined as the volume of medium that is at least three times the saturated solubility of the API, with the lowest quantity of designated surfactant. These studies allow optimization and observing the amount of surfactant that is needed to solvate the fill material within a time that is relevant to the dissolution test. It is more reasonable that a dissolution result reflects the properties of the API under the sink conditions; however, a medium that fails to provide sink conditions may be acceptable by the USP if it is appropriately justified. Likewise, when choosing the medium, the effect of additives such as acid and salt concentration, buffer counter-ions and co-solvents, and types of enzymes and their activity must also be evaluated and justified, if used. The solubility improvement of the API depends on various factors, including the nature of the surfactant and the fill material, temperature, pH, and ionic strength. This relationship should be understood for different surfactants and compounds before executing the dissolution experiment.

Typical media for dissolution studies include: dilute hydrochloric acid (0.1 N), buffers in the physiologic pH range of 1 to 7.5 (i.e., phosphate, acetate, or citrate), simulated gastric or intestinal fluid (with or without enzymes), water, and surfactants such as Tween, Brij 35, Triton, polysorbate 80, cetyl trimethyl ammonium bromide (CTAB), sodium lauryl sulfate (SLS), and bile salts [100]. Some SGC formulations may contain a matrix or API that is not soluble in water or acidic environment and consequently, does not meet sink conditions in aqueous solution. In these instances, surfactants with a justified concentration may be added to the dissolution medium. The choice of surfactant and its concentration in relation to solubility and physical stability of the API is critical and must be optimized, understood, and justified. The addition of surfactant should reflect changes in the formulation and interactions among fill components and may shed light on the in vivo behavior of the SGCs.

Surfactants play a role in dissolution by replacing water molecules on the particle surface, which reduces interfacial tension between the solution and the surface [101]. Amidon et al. has proposed that the use of media containing surfactants is a suitable method for solubilizing such drugs because various surfactants are present in the GI fluid, e.g., bile salts, lecithin, cholesterol and its esters [102]. They consist of two distinct components, hydrophilic and hydrophobic, and are categorized into four groups according to the charge on the hydrophilic group: anionic (e.g., sodium lauryl sulfate (SLS)), cationic (e.g., cetyl trimethyl ammonium bromide (CTAB), zwitterionic (e.g., alkyl betaine) [101], and non-ionic (e.g., Tween and Triton) [103,104]. Dissolution media containing cationic surfactants are better able to discriminate dissolution rates of acidic fill materials, while anionic surfactants differentiate better for basic fill materials. SLS has been reported to be the most commonly used surfactant in dissolution studies [100]. Solubility and dissolution rate enhancement by the surfactants are a function of surfactant concentration and the size of a micelle, and its stability, all of which can be related to the critical micelle concentration (CMC) [105]. The CMC is defined as the minimum concentration of a surfactant&#;s monomer at which it aggregates to micelles and is characteristic for each surfactant. A lower CMC value for a given surfactant means the micelles are more stable [106]. Furthermore, the knowledge of the molecular structure of the surfactant can provide information on the size of the micelles.

It is important to note that the addition of surfactant to dissolution media can sometimes cause a decrease in the dissolution rates of some drug products, and in some instances can also distort drug peaks during high-performance liquid chromatography (HPLC) analysis ( ). In a previous study [63], it was found that an immediate-release SGC, containing a poorly soluble drug, loratadine, showed peaks distortion in the presence of SLS. A similar observation of a decrease in the dissolution of gelatin capsules with SLS at lower pH has also been reported by other research groups [107,108].

The development of simulated fluids for dissolution testing requires understanding of the physiological conditions of the GIT. It is important to note that the GIT is complex and has a regional dependence drug absorption [109]. Several physiological factors that can affect the dissolution process in vivo include: surfactants in gastric juice and bile, viscosity of the GI contents, GI mobility patterns, GI secretions, pH, buffer capacity, and co-administration of fluids or food [110]. Vertzoni et al. [111] developed a fasted-state simulated gastric fluid (FaSSGF) containing sodium taurocholate, lecithin, and pepsin at pH of 6.5 in order to assess its importance for the in vivo dissolution of lipophilic compounds. The authors concluded that simulation of the gastric content was essential in order to assess the absorption profile of lipophilic weak bases. An overview of the composition of the common in vitro bio-relevant dissolution media is provided by Klein [112] and Galia et al. [113]. Likewise, simulated dissolution media must take into account the developmental changes in gastrointestinal fluid composition because these can result in variations in luminal drug solubility between children and adults. Therefore, evaluating age-specific changes in GI fluid parameters (i.e., pepsin concentration, bile acids, luminal viscosity, pH, osmolality, etc.) is very important in order to define the composition of bio-relevant dissolution media in pediatrics [114]. Furthermore, aged population with medical conditions such as hypochlorhydria and achlorhydria have elevated gastric pH [115]. Therefore, simulated dissolution media in this population may need to be adjusted to reflect this increased pH.

The selection of dissolution apparatus is another critical step in the dissolution evaluation of SGCs, as the mixing efficiency of fill material contents with media is very much influenced by the agitation hydrodynamics, particularly to variables such as paddle rotation speed. The two commonly used methods for evaluating the dissolution properties of SGCs are the paddle and basket methods ( ).

A basket apparatus has the advantage of enclosing SGCs. This method may be selected if SGCs are filled with a material that has a specific gravity less than that of water, where baskets prevent the SGC and its components from floating in the medium. One common problem observed using the basket is that during the dissolution experiment, the soft gel shell may disintegrate into a soft and sticky mass that can clog the basket&#;s mesh, generating high variability in the results. Additionally, if the fill material is hydrophobic, i.e., an oil-based fill, dispersion into fine droplets that can pass through the basket&#;s mesh may not take place, giving rise to a delay in dissolution that is not representative of the true properties of the SGCs. To mitigate this problem, an alternative would be using a basket with larger pores, i.e., 20 or 10 mesh sizes [116]. Pillay and Fassihi used a rotating basket method to evaluate the dissolution of lipid-based SGCs of nifedipine. Their data showed that, after six hours of dissolution test, most of the viscous oily fill formulation was still entangled within the baskets and this led to the dissolution failure [55]. This was attributed to using the standard dissolution basket with pores size of 40 mesh, combined with inappropriate hydrodynamic conditions within the basket. However, when the dissolution test was repeated using a re-designed dissolution apparatus, in this case, nifedipine SGCs showed the best dissolution profiles.

The paddle method constitutes about 70% of the dissolution methods used by FDA-approved commercial drug products [100]. This method does not use a mesh basket to contain the capsules, and so a common initial problem observed in this method is the floating of the SGCs to the surface of the dissolution medium once it breaks. In these instances, wire coils, also known as sinkers, can be used to enclose the soft gels and hold them on the bottom of the vessel [117]. This allows the fill to be better exposed to the medium (upon shell rupture) and helps to prevent the capsule from sticking to the vessel walls. The shape and size of the sinker should be selected carefully as it can impact the dissolution process, especially in cases where SGCs swell when they encounter the dissolution medium. In previous study, it was shown that the dissolution rate obtained using the paddle method was faster, highly variable at lower time points than those obtained using the basket. In contrast, the data collected using the basket dissolution apparatus showed that the method was more selective and had less variation in terms of API release profile [63]. shows examples of SGCs that are commercially available and their dissolution methods. Other research groups have evaluated the feasibility of using the USP III in evaluating the dissolution of SGCs. Monterroza and Ponce De León [118] developed a discriminating dissolution method of SGCs containing an oily suspension of micronized progesterone. They compared the dissolution profiles generated using USP 1, 2, and 3. After preliminary tests, USP 1 and USP 2 methods did not reach the target of releasing more than 85% of the API in less than 90 min. However, USP 3 showed promising prospect of releasing more than 85% of the API in less than 90 min in the presence of 250 mL of 4% of SLS in pH 6.8 phosphate.

Table 3

Drug Product InformationDissolution MethodCyclosporine (100 mg)Apparatus 2 at 75 rpm in mL 0.1 N HCl containing 4 mg of N,N-dimethydodecylamine-N-oxide per mLDutasterideTier 1: Apparatus 2 at 50 rpm in 900 mL 0.1 N HCI with 2% (w/v) SLS. Tier 2: Apparatus 2 at 50 rpm in 0.1 N HCI with pepsin (as per USP) (450 mL) for the first 25 min, followed by addition of 0.1 N HCI with SLS (4% w/v) (450 mL) for the remainder of the dissolution testIsotretinoinApparatus 1 with 20 mesh at 100 rpm in 900 mL 0.05 M Potassium Phosphate Buffer, dibasic, pH 7.8, containing 0.5% lauryldimethylamine N-oxide (LDAO)ParicalcitolApparatus 1 at 100 rpm in 500 mL in 4 mg/mL of 0.4% lauryldimethylamine N-oxide (LDAO)ErgocalciferolApparatus 2 at 100 rpm in 500 mL 0.5 N NaOH with 10% Triton-X-100Lopinavir/RitonavirApparatus 2 at 50 rpm in 900 mL, Tier 1: 0.05 M Polyoxyethylene 10 Lauryl Ether with 10 mM Sodium Phosphate monobasic (pH 6.8); Tier 2: same as above with not more than (NMT) USP units/L of PancreatinAmprenavirApparatus 2 at 75 rpm in 900 mL 0.1 N HClLoratadineApparatus 2 with sinker at 75 rpm in 900 mL. Tier 1: 0.1 N HCl with 0.1% Tween 20. Tier 2: 0.1 N HCl with 0.1% Tween 20 with addition of pepsin (as per USP)IbuprofenApparatus 1 at 150 rpm in 900 mL 50 mM Phosphate Buffer, pH 7.2Open in a separate window

In some cases, such as coated SGCs, a two-step or two-tier dissolution technique must be developed [120,121,122]. The purpose of this method is to assess the integrity of the coating in the acidic conditions of the stomach and measure the drug release in lower parts of the GIT, which have near-neutral pH conditions. Manually performing the two-step dissolution test is labor-intensive and requires well-trained analysts. For example, it requires pre-heating the second medium solution, adjusting the medium by adding the second part of the solution as well as adjusting and confirming pH for six vessels within 5 min. Typically, there are two approaches towards medium modification known as medium-addition or medium-exchange. For example, both approaches may start with an acidic step, such as 0.1 N hydrochloric acid, for a certain period, followed with a buffer step, such as phosphate buffer at pH 6.8. The specific time is chosen as needed for the individual drug product. While using either approach, the pH adjustment must be accomplished in a controlled and reproducible manner via pre-heated media. The operation of adding and adjusting the pH must be done within 5 min [123]. Zhao and co-workers described a two-step dissolution method using medium addition and paddle apparatus, in which the surfactant Tween 80 was included in the media to enhance the solubility of the API in the first stage [124]. The developed dissolution method was able to discriminate against the changes in composition, manufacturing process, and stability of the drug product. When developing a two-step dissolution procedure, several factors must be carefully examined to establish a suitable medium. The most critical step is to carefully evaluate different media to identify the one that achieves the sink conditions. The fill material may have a pH-dependent solubility, so an evaluation of the solubility of the compound in both the acidic and neutral media must be made. For instance, 0.1 N HCl and 50 mM pH 6.8 phosphate buffers are commonly used media.

The medium-addition technique, which is used for a two-step dissolution for enteric-coated capsules or two-tier dissolution testing, uses paddle or basket apparatus. This approach requires the addition of a relatively small amount of medium to each vessel in a short time. Generally, the common dissolution volumes used are in the range of 500 to mL, with 900 mL being the most commonly used in the FDA-approved drug products [100]. However, the dissolution volumes should be defined by the sink conditions. To develop a robust two-step dissolution method which can be transferred to quality control, a medium addition method is preferred where a volume of, e.g., 200 mL, can be added to 700 mL initial volume to adjust pH, and then add the surfactant, or enzyme, depending on the soft gelatin capsule drug product [124]. Furthermore, an accurate volume of the medium must be added to ensure that a volumetric error does not occur. Likewise, media addition must consider the final desired pH of the final volume. This technique is less invasive for the SGCs and is easier to conduct in a short time when running multiple batches. This approach is also less labor-intensive and allows for higher sampling throughput during the experiment run. For use in enteric-coated drug products, the API should be soluble up to the specification level in the medium of the first step to be able to detect a failure in the coating. For example, if the specification level for the first step is not more than 10% released, then this medium must be able to dissolve at least 10% of the active ingredient in the soft-gelatin capsule drug product. If the fill material is not soluble in the first-step medium, a surfactant may be added to solubilize at least 10% of the API in the fill material [124]. For use in two-tier dissolution, the fill material would require the surfactant to be present to meet solubility requirements, but also needs the enzyme to overcome the cross-linking.

For the medium-exchange approach used for enteric-coated capsules, the acid medium is drained after the first step, and a full amount of pH 6.8 buffer that has been equilibrated at similar conditions is added to the same vessel for the buffer stage. The dosage form should be undisturbed during the medium change. The complete medium replacement method resembles the medium-addition approach in that the capsules are first introduced to an acidic medium. At the end of the first step, a sample for analysis is taken, and then the dosage form is removed from the acidic conditions. Removing technique of dosage form depends on the type of dissolution apparatus. The dosage forms may be manually moved from one vessel to another. Alternatively, the entire vessel containing the acid could be removed and replaced with another vessel containing the buffer, and the dosage form is transferred to the new vessel. The quality of the SGCs dosage form is ensured by meeting the USP acceptance criteria for the acid stage, i.e., less than 10% of the API is released from the drug product during the first step of the developed dissolution technique, and therefore, the coating is considered to have passed the acid-step test. If each unit release is not less than Q + 5% for the buffer stage, then the soft gel dosage form has passed the second step of dissolution [125]. Q represents the amount of an active ingredient dissolved in the dissolution medium, expressed as a percentage of the labelled content. To overcome the challenges of manual manipulations of adding the buffer solutions and adjusting the pH during the two-step dissolution testing, other research groups have developed semi-automated dissolution systems for these measurements [125]. The media exchange technique is challenging for SGCs, especially if the capsules have softened due to the liquid exposure, soaking alone will cause some softening but may not cause the rupture of the capsule. Therefore, the transfer of the capsule or media removal without disturbing the shell may be difficult due to mechanical stress.

The European Medicines Agency (EMA) has developed its own guidance on in vitro dissolution tests for immediate-release drug products [126]. In dissolution guidance, EMA describes specifications for the quantity of active substance dissolved in a specified time, which is expressed as a percentage of API on the product label. The goal of the guidance is to set specifications to ensure batch-to-batch consistency and highlight possible problems with in vivo bioavailability. The guidance for solid immediate-release (IR) drug products from the European Pharmacopoeia (Ph. Eur. 5.17.1) has some differences compared with the FDA specifications. From a pharmaceutical perspective, the European Pharmacopoeia (Ph. Eur.) states that IR formulations should normally achieve in vitro dissolution of at least 80% of the drug substance within not more than 45 min. However, based on the USP guidance, in general, 85% or more of the drug substance should be released within 30 to 45 min.

Dissolution methods for SGCs must also consider the aspect of age-related gelatin cross-linking influencing the dissolution performance. The USP <711> permits the use of a two-tier assessment of hard and SGCs when evidence of cross-linking is present. Evidence of cross-linking usually occurs based on visual observations during the performance of the dissolution testing. This is based on the fact that the USP general chapters on dissolution <711> as well as disintegration and dissolution of dietary supplements <>, allow the addition of various enzymes based on pH of the dissolution medium when hard or SGCs and gelatin-coated tablets do not conform to the dissolution or to resolve potential cross-linking issues specifications [127]. Cross-linking evidence can come in the form of poorly dissolving gelatin shell or pellicle formation, which appears as a sac surrounding and containing the fill material after the shell is dissolved (see Section 8). To overcome cross-linking, the two-tier dissolution test would involve the addition of proteolytic enzymes such as pepsin, papain, bromelain, or pancreatin to the dissolution media and repeating the dissolution [128]. These enzymes effectively digest the peptide bonds between the amino acids making up the gelatin strands in the shell. The use of enzymes for dissolution must be done with care, as the enzymes require significant mechanical mixing to get into solution, are minimally stable in solution, and can be impacted by other components of the media, such as surfactants. If a protein denaturing surfactant [129] is used in the media, a two-step tier 2 method must be performed. The first step involves the dissolution of the capsule shell using media containing an enzyme and no surfactant as a pre-treatment step. After the capsule shell is dissolved, media containing surfactant is added to complete the dissolution and solubilization of the fill and active pharmaceutical ingredient. It was observed that using the digestive enzyme while conducting the dissolution study and afterward using the surfactant showed a better effect in the two-tier method [130].

Another important aspect that is worth discussing regarding dissolution of SGCs is the concept of an in vitro&#;in vivo correlation (IVIVC). This is normally used to establish a relationship between an in vivo response (e.g., amount of drug absorbed) and an in vitro physicochemical property of a dosage form. The main objective of this concept is to make sure that the in vitro properties of two or more batches of the same drug product are performing similarly under in vivo conditions. Hence, this relationship is essentially important in guiding drug development and drug approval processes that are designed to mimic the in vivo drug release. There have been various studies on IVIVC of SGCs and some have shown good correlations. Meyer et al. [53] assessed whether the changes in the in vitro dissolution of hard and soft gelatin acetaminophen capsules, as a result of gelatin cross-linking, are predictive of changes in the bioavailability of the capsules under in vivo conditions. Their data showed that the in vitro rate of dissolution of hard and SGCs decreased due to cross-linking. On the other hand, the bioequivalence studies showed that both hard and SGCs, which failed to meet the USP dissolution specification in water, but complied when tested in SGF containing pepsin, were bioequivalent to the unstressed control capsules. Based on the plasma concentration parameters, the capsules that were cross-linked to the greatest extent were not bioequivalent with the unstressed control capsules. In another study, Nishimura et al. [131] attempted to predict the human plasma drug concentrations of SGCs containing a poorly soluble drug, arundic acid. SGCs were stored at short- and long-term conditions, i.e., 15 °C for 3 months and 25 °C (60% relative humidity (RH)) for 30 months, respectively. The authors showed that the in vitro dissolution data obtained with the dissolution medium containing surfactant (i.e., 2% SLS, pH 6.8) were more effective in predicting the drug plasma concentrations following oral administrations of the SGCs under both storage conditions. Likewise, Rossi et al. [132] developed and validated a dissolution test for ritonavir SGCs based on human in vivo pharmacokinetic data. The authors used a USP II method with 900 mL of dissolution medium containing water with 0.3%, 0.5%, 0.7%, or 1% (w/v) of SLS at rotation speed of 25 rpm. Their data showed strong level A correlation between the percent of the drug dissolved versus percent absorbed. Significant in vitro&#;in vivo correlation was achieved using dissolution medium containing water with 0.7% SLS. In another similar study, Donato et al. [133] reported similar results on the development and validation of a dissolution test for lopinavir, a poorly water-soluble drug, in soft gel capsules, based on in vivo data. In this work, a new formulation of lopinavir was developed and its dissolution tests validated using in vivo data. All formulations were evaluated for in vitro dissolution containing 2.3% SLS at pH 6.0 and USP 1 at 25 rpm. At these conditions, the authors showed strong level A correlations for the fraction dissolved versus fraction absorbed.

Understanding the IVIVC relationship of new formulations of SGCs will help the design of optimized dissolution methods that can be used to predict in vivo performance.

About Gelatin Capsules and the importance of its hardness ...

About Gelatin Capsules and the importance of its hardness testing

What is a Gelatin Capsule?

Gelatin capsules, informally called gel caps or softgel capsules, are composed of gelatin manufactured from the collagen of animal skin and bone.

There is another type of capsule known as Vegetable capsules. They are made up of cellulose. The main ingredient of vegetarian capsules is hydroxypropyl methyl cellulose (HPMC). However, in the current market, gelatin capsules are more universally used than vegetarian capsules because the cost of production is lower.

Types of Gelatin Capsules:

Hard shell Capsules: They contain dry, powdered ingredients or miniature pellets. These are made in two halves: a smaller-diameter &#;body&#; that is filled and then sealed using a larger-diameter &#;cap&#;.

Soft shell Capsules: They contain oils or active ingredients that are dissolved or suspended in oil.

Finished product Quality Control tests for Capsules:

Finished capsules are subjected to a number of tests in accordance with compendial standards and regulatory requirements for unit dose capsule products. These batteries of tests help identify whether the batch is acceptable for marketing or its intended usage

Physical Integrity of Gelatin Capsules:

Leaking gelatin capsules destroys consumer confidence in the product and the manufacturer. To prevent defective capsules from reaching the market, a manufacturer must develop tests to identify them. One approach is to use a Gelatin capsule hardness tester that applies a compressive force to gelatin capsules to confirm they have sufficient wall strength to withstand external forces during manufacturing, storage, packaging, transport and finally when used by a consumer.

What should the hardness be of a Gelatin capsule?

In general, a suitable weight ratio is that dry plasticizer: dry gelatin = 0.4 &#; 0.6: 1.0, and the ratio of water to dry gelatin is 1:1. The capsule shell is the hardest when the weight ratio between the plasticizer and gelatin is 0.3:1.0, and the shell is soft if 1.8:1.

What is a Gelatin Capsule Hardness Tester?

Gelatin Capsule Hardness tester is a device that indicates the hardness of a capsule by measuring the effect on its surface of a localized penetration by a standardized flat shaped indenter made of hard steel.  Bareiss has made an exclusive solution to test the hardness of a capsule called a Gelomat

For more information, please visit Cell Instruments.

Application of Gelomat and its importance:

Gelomat is a plug and play hardness tester and non-destructive method developed for the determination of the hardness and resistance on gelatin, gelatin capsules, play dough, paintballs, agars etc.

The unique design and digital measuring systems ensure that a remarkably high measuring precision and most reliable measurement is obtained. An automatic measurement for hardness test on gelatin capsules is required to obtain optimum accuracy and repeatability.

The user can choose a measuring head from either 0 &#; 2 N or 0 &#; 20 N and has an option to attach an Automatic positioning device called Rotofix or a manually operated sample fixture known as

Centrofix. With the help of our software, users can analyze the results, store data, see histograms, create batch folders, etc.

Integration with an Automated optical inspection system:

Based on the client&#;s requirements, Bareiss also designs and develops customized automated optical inspection systems that could be integrated with a Gelomat hardness tester or as a stand alone. This system will allow the quality specialists to check the size and defects of the finished product, achieving close to 100% perfection in quality for each batch that is manufactured.  This efficient technology will maximize your business values and at the same time embrace cost optimization for your company.

 

Gelatin Capsules and Its Importance In Hardness Testing

 

 

 

Gelatin capsules or gel caps or softgel capsules, are composed of gelatin manufactured from the collagen of animal skin and bone.

There are Vegetable capsules as well made up of cellulose. The main ingredient of vegetarian capsules is hydroxypropyl methyl cellulose (HPMC).

However, in the current market, gelatin capsules are more extensively used than vegetarian capsules  because the cost of production is lower.

Types of Gelatin Capsules:

Hard shell Capsules contain dry, powdered ingredients or miniature pellets. These are made in two halves: a smaller-diameter &#;body&#; that is filled and then sealed using a larger-diameter &#;cap&#;.

Soft shell Capsules contain oils or active ingredients that are dissolved or suspended in oil.

Finished capsules are subjected to a number of tests in accordance with compendial standards and regulatory requirements for unit dose capsule products. These tests help identify whether the batch is acceptable for marketing or its intended usage.

Leaking gelatin capsules destroy consumer confidence in the product and the manufacturer. To prevent defective capsules from reaching the market, a manufacturer must develop tests to identify them.

Gelatin Capsule Hardness tester is a device that indicates the hardness of a capsule by measuring the effect on its surface of a localized penetration by a standardized flat shaped indenter made of hard steel.  Bareiss has made an exclusive solution to test the hardness of a capsule called a Gelomat.

Gelomat is a plug and play hardness tester and non-destructive method developed for determination of the hardness and resistance on gelatin, gelatin capsules, play dough, paintballs, agars, etc.

The unique design and digital measuring systems ensures that a very high measuring precision and most reliable measurement is obtained. An automatic measurement for hardness test on gelatin capsules is required to obtain optimum accuracy and repeatability.

The user can choose a measuring head from either 0 - 2 N or 0 - 20 N and has an option to attach an Automatic positioning device called Rotofix or a manually operated sample fixture known as Centrofix. With the help of our software, user can analysis the results, store data, see histograms, create batch folders, etc.

 

The Vital Role of Gelatin Capsule Hardness in Pharmaceutical Manufacturing

Unveiling the importance of gelatin capsule hardness tests, including softgels, for maintaining pharmaceutical standards with Cell Instruments CHT-01 Gelatin Capsule Hardness Tester.

Delve into the significance of hardness tests for gelatin capsules in pharmaceuticals, discover their impact on quality, dosage accuracy, and learn about Cell Instruments CHT-01&#;s role in ensuring regulatory compliance.

Gelatin Capsule Hardness: Cornerstone of Pharmaceutical Quality

The Critical Nature of Hardness Testing for Gelatin Capsules

Gelatin capsule hardness, encompassing hard capsules and softgels, remains integral to the pharmaceutical industry&#;s quality assurance. This detailed guide explores the necessity of hardness testing in certifying that capsules adhere to stringent industry criteria, thereby affirming the medicines&#; efficacy, safety, and dependability.

Gelatin Capsules in Drug Delivery: Hard Capsules vs. Softgels

Recognized for their role in drug encapsulation, gelatin capsules are available in two major forms. Their hardness is vital for the drug&#;s quality, affecting everything from manufacturing to patient administration.

The Process of Assessing Capsule Hardness

The resistance of a capsule&#;s shell against deformation is what hardness testing aims to measure, using tools like the Cell Instruments CHT-01. This assessment is crucial for confirming the material&#;s robustness.

Capsule Hardness Testing: Beyond Quality Assurance

Upholding Product Integrity Through Hardness Testing

Capable of influencing drug stability and efficacy, capsule hardness helps ensure product consistency. Through meticulous hardness evaluations, manufacturers address strength-related issues, upholding high-quality standards and regulatory mandates.

Ensuring Accuracy in Medication Dosage

For accurate drug delivery, consistent capsule hardness is vital. Deviations can affect drug release, patient safety, and therapeutic efficiency. Proper hardness guarantees each capsule conveys the correct medication dosage.

Encouraging Patient Adherence with Optimal Capsule Properties

The hardness of a capsule can impact patient compliance. Capsules with suitable hardness levels facilitate an improved patient experience, thus enhancing their adherence to medication schedules.

Compliance with Regulatory Guidelines and GMPs

Adherence to regulations like the FDA and EMA is obligatory, with hardness testing being a regulatory focus. Adhering to GMPs and performing thorough hardness assessments, manufacturers showcase a dedication to product quality and compliance.

Optimizing Manufacturing Through Hardness Testing

Hardness evaluations also aid in refining manufacturing processes. Monitoring this attribute helps identify production inconsistencies and implement timely solutions, improving operational efficiency and cost-effectiveness.

 

A Leap Forward in Capsule Hardness Technology

Featuring advanced sensors, software, and user interfaces, the CHT-01 capsule hardness tester exemplifies modernization in quality control measures. This instrument augments precision across hardness testing protocols.

Probing Deeper: Compression and Penetration Testing with the CHT-01

The CHT-01 leverages cutting-edge methodologies like compression and penetration testing to evaluate the hardness of both hard and softgel capsules, detailing their durability and resistance against various pressures.

Data Integration and Easy Operation with the CHT-01 Interface

With its easy-to-navigate software, theCHT-01 Gelatin Capsule Hardness Tester aligns with other quality control systems, enhancing data analysis and reporting procedures, supporting comprehensive test documentation.

The Cell Instruments CHT-01 Gelatin Capsule Hardness Tester

In essence, gelatin capsule hardness testing forms a pivotal part of the drug manufacturing process, guaranteeing the uprightness, effectiveness, and security of medications encapsulated within gelatin capsules. The Cell Instruments CHT-01 Gelatin Capsule Hardness Tester. is instrumental in refining quality control operations. It aligns with industry regulations, facilitating the delivery of superior-grade pharmaceuticals. Its advanced functionalities and ease of operation render it an essential advancement in capsule hardness testing.

 

Shopper satisfaction is our primary focus. We insist on the same level of professionalism, quality, reputation and maintenance of Automatic Capsule Hardness Tester,Medical Device Tester,Medical Product Testing Instruments Manufacturer,Motorized Cap Torque Tester,Container Tester,Fabric Thickness Tester,Digital Test Stand,Vertical Tensile Tester

If you want to learn more, please visit our website gelatin capsule hardness tester.