Differences in In Vitro Dissolution Rates Using Single-Point and Multi-Point Sampling

Similar documents
Practical Pharmaceutical Technology I USP Dissolution Method for PARACETAMOL 500 mg Tablets Section No. 6 Group D

Scholars Research Library

Development of Discriminating Method for Dissolution of Aceclofenac Marketed Formulations

To assess product performance and the quality of

Opportunities for Regulatory Relief via In Vitro Dissolution. James E. Polli June 10, 2015

Enteric coating is a widely used technique for

Available online through ISSN

An Automated Application Template for Dissolution Studies

King Saud University College of Pharmacy Department of Pharmaceutics. Biopharmaceutics PHT 414. Laboratory Assignments 2010 G 1431 H

Scientific Principles and Advanced Concepts in Dissolution

DETERMINATION OF DRUG RELEASE DURING DISSOLUTION OF NICORANDIL IN TABLET DOSAGE FORM BY USING REVERSE PHASE HIGH PERFORMANCE LIQUID CHROMATOGRAPHY

DISSOLUTION RATE LIMITING AUC: SIMPLE METHODOLOGY FOR MEASURING DISSOLUTION RATE OF THE ENTIRE DOSE IN BIORELEVANT MEDIA

Fasted State Dissolution Protocol

Research Article. Dissolution Study of Oxolamine Citrate by UV Spectrophotometric Method in Pharmaceutical Dosage Form

CDER Risk Assessment to Evaluate Potential Risks from the Use of Nanomaterials in Drug Products

Comparison of US Pharmacopeia Simulated Intestinal Fluid TS (without pancreatin)

Setting Attainable and Practical Particle Size Specifications

LABORATORY EXERCISE: USING SPECTROPHOTOMETRY FOR QUALITY CONTROL: NIACIN

ANALYTICAL METHOD PROCEDURES

STUDY OF THE APPLICABILTY OF CONTENT UNIFORMITY AND DISSOLUTION VARIATION TEST ON ROPINIROLE HYDROCHLORIDE TABLETS

FACULTY OF PHARMACY. M. Pharmacy I Semester (Suppl.) Examination, November 2015 (Common To All) Subject: Pharmaceutical Analytical Techniques

ActiPix SDI300 Surface Dissolution Imaging System

Plop Plop, Fizz Fizz, Oh What A Relief It Is (Which Pain Reliever Works Fastest)

Dissolution testing is considered as an important tool

Solubility and Dissolution Rate Determination of Different Antiretroviral Drugs in Different ph Media Using UV Visible Spectrophotometer

Chapter 4: Verification of compendial methods

International Journal of Pharmacy and Pharmaceutical Sciences Vol 2, Issue 1, 2010

Journal of Pharmaceutical and Biomedical Analysis Letters. Analysis Letters

Monitoring Dissolution Rate of Amiodarone Tablets by a Multiple Fiber-Optic Sensor System

Dissolution Method Development and Validation of Paracetamol Aceclofenac Tablets

The Nitrofurantoin Capsules Revision Bulletin supersedes the currently official monograph.

Plop Plop, Fizz Fizz, Oh What A Relief It Is (Which Pain Reliever Works Fastest)

International Journal of PharmTech Research CODEN (USA): IJPRIF, ISSN: , ISSN(Online): Vol.9, No.7, pp , 2016

USP Method Transfer and Routine Use Analysis of Irbesartan Tablets from HPLC to UPLC

IMPROVEMENT OF DISSOLUTION PROFILE OF LORNOXICAM BY SOLID DISPERSION USING SPRAY DRYING TECHNIQUE

IVIVC Industry Perspective with Illustrative Examples

Formulation of Low Dose Medicines - Theory and Practice

Revision Bulletin 27 Jan Feb 2017 Non-Botanical Dietary Supplements Compliance

In Vivo Predictive Dissolution Flux Measurements. Konstantin Tsinman, PhD Chief Scientific Officer Pion Inc

104 Full Text Available On Research Article!!! Pharmaceutical Sciences. Received: ; Accepted:

Revision Bulletin 29 Dec Jan 2018 Non-Botanical Dietary Supplements Compliance

Christin T. Choma TA Instruments, 109 Lukens Drive, New Castle, DE 19720, USA

Ketorolac tromethamine (KT)[1] is

Test Method Development and Validation as Pertaining to Microtrac Particle Size Measuring Instruments

Journal of Chemical and Pharmaceutical Research, 2012, 4(6): Research Article. Estimation of zaleplon by a new RP-HPLC method

In the present study, parameters such as solubility, medium ph, surfactant type and dissolution behavior of formulations,

CHAPTER - 3 ANALYTICAL PROFILE. 3.1 Estimation of Drug in Pharmaceutical Formulation Estimation of Drugs

VALIDATION OF A UPLC METHOD FOR A BENZOCAINE, BUTAMBEN, AND TETRACAINE HYDROCHLORIDE TOPICAL SOLUTION

Application Note PB 401. Improve HPLC Sample Preparation in an Analytical Laboratory A New Automated Sample Preparation Process

Independence and Dependence in Calibration: A Discussion FDA and EMA Guidelines

Validated First Order Derivative Spectroscopic Method for the determination of Stavudine in Bulk and Pharmaceutical Dosage Forms

Preparation And Characterization Of Simvastatin Nanosuspension By Homogenization Method

7. Stability indicating analytical method development and validation of Ramipril and Amlodipine in capsule dosage form by HPLC.

Hydrodynamic Complexity Induced by the Pulsing Flow Field in USP Dissolution Apparatus 4

Spectroscopic Method For Estimation of Atorvastatin Calcium in Tablet Dosage Form

To control the consistency and quality

KHAN HAJERA*, MIRZA SHAHED. Y.B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Rauza Bagh, Aurangabad

Additionally, minor editorial changes have been made to update the monograph to current USP style.

DISSOLUTION TEST FOR SOLID ORAL DOSAGE FORMS. Proposal for revision for The International Pharmacopoeia. (February 2018)

Reverse Phase High Performance Liquid Chromatography method for determination of Lercanidipine hydrochloride in bulk and tablet dosage form

Received: ; Accepted:

Measurement of Intrinsic Drug Dissolution Rates Using Two Types of Apparatus

Validated spectrophotometric determination of Fenofibrate in formulation

Research Article Spectrophotometric Estimation of Didanosine in Bulk Drug and its Formulation

Physicochemical Characterization of Acyclovir Topical Semisolid Dosage Forms Towards TCS Validation Flavian Ștefan Rădulescu, Dalia Simona Miron

DEVELOPMENT AND VALIDATION OF HPLC METHOD OF DISSOLUTION TEST FOR METOPROLOL SUCCINATE AND CILNIDIPINE

BASIC CONCEPTS C HAPTER 1

[ 11 C]NNC 112 FOR INJECTION: CHEMISTRY, MANUFACTURING AND CONTROLS

Monitoring Emulsion Polymerization by Raman Spectroscopy

The Influence of Hydro-Alcoholic Media on Hypromellose Matrix Systems

Method Development and Validation Of Prasugrel Tablets By RP- HPLC

Aripiprazole tablets: Development and Validation of a Dissolution Methodology

Journal home page:

Volume: I: Issue-2: Aug-Oct ISSN

Dissolution Automation: Basic Questionnaire.

Dissolution study and method validation of alprazolam by high performance liquid chromatography method in pharmaceutical dosage form

Simultaneous UV Spectrophotometric Method for the Estimation of Cefuroxime Axetil and Probenecid from Solid Dosage Forms

IN VITRO DISSOLUTION KINETIC STUDY OF THEOPHYLLINE FROM HYDROPHILIC AND HYDROPHOBIC MATRICES

Intercontinental journal of pharmaceutical Investigations and Research

TABLE OF CONTENTS. vii

Hydrodynamics are a commonly considered

Supplementary Material

Simultaneous Estimation of Residual Solvents (Isopropyl Alcohol and Dichloromethane) in Dosage Form by GC-HS-FID

Quality by Design in the Development of Analytical Procedures

Journal of Drug Delivery and Therapeutics

Dissolution Test 5 was validated using a Zodiac C18 brand of L1 column. The typical retention time for atorvastatin is about min.

DI 3500 Discrete Aoalvzer

RP-HPLC Method Development and Validation of Dapagliflozin in Bulk and Tablet formulation

contents of the currently official monograph. Please refer to the current edition of the USP NF for official text.

Revision Bulletin Official April 1, 2014 Alprazolam 1

22 Oral Solid Dosage Forms Chapter Objectives At the conclusion of this chapter the student should be able to:

Mixed Hierarchical Models for the Process Environment

Development and Validation of Stability Indicating RP-HPLC Method for the Determination of Anagrelide HCl in Pharmaceutical Formulation

Research Article. UV Spectrophotometric Estimation of Alprazolam by second and third order derivative Methods in Bulk and Pharmaceutical Dosage Form

SIMPLE AND SENSITIVE VALIDATED REVERSE PHASE HPLC-UV METHOD FOR THE DETERMINATION OF LYMECYCLINE IN PHARMACEUTICAL DOSAGE FORMS

ASEAN GUIDELINES FOR VALIDATION OF ANALYTICAL PROCEDURES

Statistics and modeling in in vitro release studies

Implementation of Methods Translation between Liquid Chromatography Instrumentation

USP 36 Official Monographs / Metformin carding the first 3 ml of filtrate. Transfer 25 ml of the Analysis

Transcription:

dx.doi.org/10.14227/dt140407p27 Differences in In Vitro Dissolution Rates Using Single-Point and Multi-Point Sampling e-mail: Limin.Zhang@bms.com Limin Zhang 1, 3, Khanh Ha 1, Brent Kleintop 1, Shannon Phillips 2, Barry Scheer 1 1 Analytical Research & Development, Bristol-Myers Squibb Pharmaceutical Research Institute, New Brunswick, NJ. 2 The Dow Chemical Company, Texas Operations, Freeport, TX. INTRODUCTION In vitro dissolution testing is an important physicochemical tool used to measure drug release rates during both early and late stages of drug development. In both cases, a product-specific discriminating dissolution method is utilized as a quality control tool to evaluate batch-to-batch consistency and as a predictive means to gain a post-market bio-waiver in cases where there is in vitro in vivo correlation or in vitro in vivo relation. Developing rugged methods with appropriate discriminatory dissolution parameters is often a challenging objective regardless of the solubility of the compounds. Method development is further challenged when hydrodynamic effects within the vessel (1 4) impact method ruggedness. The hydrodynamic environment within the vessel can be affected by many variables including the position of the tablet (5, 6) and the presence and position of a measuring (7) or sampling probe (8). Thus, for poorly soluble drugs, it is challenging to develop an appropriate dissolution method that is rugged, discriminating, and maintains sufficient sink conditions. While implementing a dissolution method in a manufacturing quality control (QC) laboratory, we encountered a unique method ruggedness issue. Previously, we successfully transferred the validated discriminating dissolution method from our R&D facility to both a stability laboratory and a manufacturing QC laboratory. However, during the manufacturing validation campaign, several tablet batches exhibited slower dissolution rates in the QC laboratory when compared with the historical dissolution rates generated in the R&D and stability laboratories. Several of the process validation batches were at risk of proposed specification failure, which had required Stage II (S 2 ) testing. An investigation was initiated to determine if the root cause of the low results was due to a method robustness or product quality issue. The drug product that exhibited this behavior was prepared using a BCS class II (low solubility and high permeability) compound in which dissolution was the 3 Corresponding author. rate-limiting step for the in vivo absorption. During development and stability assessment of the drug product, in vitro drug release was routinely evaluated using a typical dissolution profile obtained by sampling at several discrete time points. Batch-to-batch variability was assessed by determining the statistical difference between the dissolution profiles of each batch (f 2 similarity test). The best performing (most discriminatory) batches against which the other batches were compared, were designated based on the dissolution profiles of prototypes with different types of granulation, type or amount of excipients, and batch chemical stability. During the process validation campaign, dissolution testing was conducted using a single sample pull at one specified time point to meet an acceptance criterion for batch release. Since some of the batches required S 2 testing, an initial investigation was focused on the effects that sampling technique had on the hydrodynamics of the dissolution vessel, such as manual versus automated sampling, or single-point versus multi-point profiling. In addition, other method parameters such as paddle rotation speed and media ph were evaluated as part of this investigation. EXPERIMENTAL This investigation was performed on a tablet dosage form containing a weakly basic BCS Class II (low solubility and high permeability) drug compound, with a pk a value of 3.6. All batches listed in this investigation were made from the same or similar formulation and manufacture processes. The dissolution method under investigation employed compendia conditions with USP Apparatus 2 (paddle) at a rotation speed of 50 rpm in a medium of 50 mm, ph 6.8 phosphate buffer. The discriminatory ability of these method conditions was agreed upon after an FDA-interface meeting prior to filing the NDA for this product. The time points for multi-point (profile) sampling used to support all phases of clinical development and stability were 10, 20, 30, 45, and 60 min. Single-point sampling at 30 min was used for release testing in the QC laboratory. Quantitation of the dissolution samples was performed by HPLC with UV detection. 27 diss-14-04-08.indd 27 11/8/2007 1:57:47 PM

The automated dissolution method used to support all phases of clinical development, technology transfer, clinical stability, and registration stability utilized a Multidose G3 manufactured by Caliper Science, Inc. This automated system can perform several unattended cycles of media dispensing, temperature equilibration, vessel filling, temperature reading, sample drop, dissolution, sampling, filtration, sample dispensing, and vessel washing. RESULTS AND DISCUSSION The registration dissolution method employed a dissolution medium of ph 6.8 based on the physiologic ph, sink condition, and acceptable solubility. As shown in the aqueous solubility curve for the subject drug compound (Figure 1), the solubility changes significantly within the ph range of 6.5 7.2, suggesting that small changes in the dissolution media ph within this range could lead to significant differences in the dissolution profile as sink conditions are approached. At the ph of 6.8 used in the method, the solubility of the active drug is 44 µg/ml. For a tablet with 2.5-mg label strength, the dissolution concentration (2.5 µg/ml) corresponds to a sink condition that is 17 times the saturation volume. Figure 1. ph-solubility profile for the subject drug compound (dashed line indicates the ph utilized by the method). Aqueous solubility at 37 C is 32 µg/ml at ph 6.7, 44 µg/ml at ph 6.8, and 71 µg/ml at ph 6.9, respectively. However, since solubility is a primary driver for the dissolution of this drug substance, method robustness could be affected by several factors such as ph or hydrodynamic disturbances. During validation of the manufacturing process for this tablet product, dissolution testing of batches was performed by the QC laboratory using automated single-point sampling at the 30-min time point. As shown in Table 1, several batches exhibited low dissolution rates that failed the Stage I specification of 80% (Q) dissolved in 30 min, resulting in the need to initiate S 2 testing. Understanding the root cause of these low results became essential since it could be indicative of a method ruggedness or product quality issue. Additionally, it is undesirable to incur a high frequency of S 2 testing from a product quality, manufacturing process robustness, and operational efficiency perspective. During the investigation of the S 1 dissolution failures, the tablet batches were reanalyzed by the R&D laboratory using an automated profile method. As shown in Table 2, the R&D laboratory dissolution results were 6 7% higher than those generated in the QC laboratory using the automated single-point method. These higher results were consistent with our previous development experience. This discrepancy between the R&D and QC laboratories was not observed during the interlaboratory method transfer, where profile sampling was performed in both laboratories (Table 3). Because the sampling technique (single-point versus multi-point profiling) was the only major difference in the procedure used by the two laboratories, sampling effects became the main focus of subsequent investigations. Comparison of Dissolution Data Using Single-Point versus Multi-Point Sampling A comparison of single-point and multi-point sampling across different automated dissolution systems from multiple laboratories showed a consistent 3 5% bias (Table 4). The consistency of the bias suggested that the observed difference was inherent to the method parameters and was not a function of the instrument, site, and analyst. It further suggested that the automated dissolution systems were functioning properly across all laboratories and that method reproducibility was not an issue. Table 1. Dissolution Results for Process Validation Batches Using Single-Point Sampling. Percent of Label Dissolved at 30 min * Tablet #1 Tablet #2 Tablet #3 Tablet #4 Tablet #5 Tablet #6 Average Batch 1 90 87 88 83 85 85 86 Batch 2 87 88 88 86 88 82 87 Batch 3 87 87 86 85 86 83 86 28 * Specification: Not less than 80%(Q) of label dissolved in 30 min. Figures in bold represent results that failed the proposed specification of 80% (Q) +5% at Stage I. diss-14-04-08.indd 28 11/8/2007 1:57:47 PM

Table 2. Comparison of Dissolution Results from Process Validation Batches Between AR&D (Multi-Point Sampling) and QC Laboratories (Single-Point Sampling). Percent of Label Dissolved at 30 min (n=12) Average (AR&D) Average (QC) Difference Batch 1 93 86 7 Batch 2 93 87 6 Batch 3 93 86 7 Table 3. Dissolution Data Generated During Method Technology Transfer Using Automated System. Average Percent of Label Dissolved (n=12) * Transferring Lab Receiving Lab Receiving Lab #2 Time Point (min) (AR&D) #1 (QC) (Stability) 10 80 81 82 20 94 93 92 30 96 95 94 45 98 96 96 60 99 96 96 * Only profile sampling was performed during method transfer. Results from all labs were within acceptance criteria. Results in bold represent the results of 30-min time point. A representative clinical batch was used for technology transfer. Table 4. Comparison of Single-Point and Profile Dissolution Results Obtained Using Multiple Automated Systems. Average Percent of Label Dissolved at 30 min (n=12) * Automated System # Single-Point Sampling Multi-Point Sampling Difference 1 89 94 5 2 93 96 3 3 86 91 5 Re-Evaluation of the Validation Parameters of the Automated Dissolution Method The slower dissolution rates were observed with both the manual and automated methodologies when single-point sampling was performed, as shown in Figure 2. For thoroughness, we explored several potential sources of sample loss due to the unique features of the automated system versus the manual system sampling. Tubing flush volume and filter loading were among the validation parameters that were reevaluated, since they could potentially contribute to loss of drug. Results indicated that these sampling parameters were properly Figure 2. Comparison of results obtained for single-point and multi-point sampling at 30-min time point on a representative clinical release batch. A 5 6% faster release was obtained when using multi-point sampling. validated and did not contribute significantly to any drug loss. Effect of Sampling Probe The automated sampling mechanism consisted of a sample pump capable of draw speeds from 5 to 12 ml/min. The sampling probe has an outer diameter of approximately 0.25 in. and a length of 9 in. The sampling position was automatically adjusted to meet USP requirements for a particular medium volume. In order to eliminate sample carryover, a sample volume of 16 ml was required. To collect this volume with the pump set at the maximum speed of 12 ml/min, the sampling probe was immersed in the dissolution medium for approximately 90 s during each sampling interval. During multi-point sampling, prior to the 30-min time point, the probe was immersed in the dissolution medium at each of the 10-min and 20-min time points for a period of 90 s. Although relatively brief, this probe immersion time could potentially cause disruption of currents within the vessel and introduce turbulence in the flow. During single-point sampling, the probe is not in contact with the medium until it withdraws the single sample at the 30-min time point. The profile sample withdrawals increase disruption of the hydrodynamic flow in the dissolution vessel, resulting in faster release of the drug at the 30-min time point as compared with single-point sampling. To determine the hydrodynamic disturbance introduced by multi-point sampling as compared with single-point sampling, it would be ideal to simulate the automated multi-point sampling by immersing the probe in the vessel without sampling for the 10-min and 20-min time points. Due to the limitation of the software, the automated dissolution system does not have the capability to sample this way. Instead, we performed a 29 diss-14-04-08.indd 29 11/8/2007 1:57:48 PM

single-point dissolution experiment where the probe was immersed in the dissolution vessel for the entire 30 min prior to sampling at the 30-min time point to determine the maximum effect this hydrodynamic disturbance would produce. If the hydrodynamic effect caused by the sampling probe was indeed the root cause of the discrepancy between single-point and profile sampling, this experiment would simulate the maximum difference that would be expected due to the presence of the sampling probe. Results in Table 5 demonstrate that at the 30-min time point, the percent drug released when the probe was immersed in the media for the entire 30 min was 6% greater than when the probe was not immersed in the media prior to the sampling at 30 min. This difference is similar to the differences observed in the previous comparison experiments, as shown in Table 2. These results suggest that the presence of the sampling probe in the media had a significant impact on the hydrodynamic environment and likely contributed to the differences in dissolution results obtained between single-point and multi-point sampling. This hydrodynamic effect was also evaluated using manual sampling for single-point and profile tests. These manual results show a similar trend when compared with automated systems (Figure 2). This trend is evident even though the cannula immersion time was much shorter for the manual method and the cannula (outer diameter of approximately 0.125 in.) is smaller than the automated probe. Since the same trend was observed in the manual system, the method appears highly susceptible to hydrodynamic effects. Evaluation of Paddle Speed and ph To further evaluate dissolution parameters that minimize hydrodynamic effects but still provide sufficient discrimatory power, experiments were conducted using different paddle speeds and media ph. The initial dissolution method submitted with the IND for this product employed a ph 7.0 phosphate buffer and 60-rpm paddle rotation speed. Due to the higher solubility of the drug at ph 7.0 and the greater agitation, this method provided a more rapid and robust release than the compendial method conditions. The results, summarized in Table 6, demonstrate that the hydrodynamic effects are minimized when higher rotation speeds and improved sink conditions (such as increased ph) are applied, Table 6. Comparison of Dissolution Results for Single-Point Versus Profile Sampling Using Altered Media ph and Paddle Rotation Speeds. Average Percent of Label Dissolved at 30 min * Single-Point Multi-Point Medium ph Paddle Speed Sampling Sampling Difference 6.8 50 RPM 86 93 91 96 3 5 6.8 60 RPM 94 95 1 7.0 50 RPM 94 96 2 7.0 60 RPM 95 96 1 regardless of the sampling technique. These conditions create a more robust dissolution method. Furthermore, the more robust dissolution method is able to discriminate the batches based on the amount of disintegrant used in the formulation, as shown in Figure 3. CONCLUSIONS Here we provide an example where hydrodynamic effects can potentially influence in vitro dissolution results. When developing a discriminating and robust dissolution method, one should evaluate how the method will ultimately be implemented in the manufacturing environment. A seemingly robust and discriminating method can present unexpected results based on the number of sampling time points and sample withdrawals. In the case of this particular drug product, it was determined that multiple sample pulls from the dissolution vessel, as in a profile experiment (typically used in the R&D or stability testing environments) versus a 30 Table 5. Comparison of Single-Point Results Obtained Using Different Sampling Strategies. Average Percent of Label Dissolved at 30 min (n=12) * Probe immersed in the Probe immersed in the vessel vessel for entire 30 min only at the 30-min time point Difference 94 88 6 Figure 3. Dissolution profiles of two prototype formulations using altered medium ph (ph 7.0) and paddle rotation speed (60 rpm). Prototype formulations containing 0% and 5% sodium croscarmellose (NaCC) were evaluated. diss-14-04-08.indd 30 11/8/2007 1:57:49 PM

single-pull (typically used in the QC environment), resulted in different dissolution rates. It is believed that this discrepancy is a result of a greater disturbance of the fluid hydrodynamics in the dissolution vessel caused by the additional insertions and residence of the sampling probe in the vessel during multi-point sampling. Additional examples in the literature suggest that the hydrodynamic conditions, drug release pattern, or mechanical forces of an in vitro dissolution are crucial to the drug release rate (9, 10). In the case of this dissolution method, the impact of the sampling technique on fluid hydrodynamics and the subsequent effect on method robustness should not be overlooked. We initially proposed a dissolution method that employed a rotation speed of 60 rpm and a medium ph of 6.8 for use during registration stability. Based on the f 2 calculation, these proposed parameters did not result in significantly different dissolution profiles as compared with the compendia conditions of 50-rpm paddle speed with a ph 6.8 medium. We chose to utilize the 50-rpm paddle speed to be in alignment with the regulatory agency suggestion. However, the hydrodynamic effects caused by sampling were not evaluated, and the revised method was found to be more sensitive to subtle changes in the dissolution conditions, such as sampling over multiple time points. Use of single-point versus multi-point profile testing should be taken into account early in the method development process. Technology transfer using single-point sampling is critical. Based on the lessons learned on this project, dissolution methods for other drug products in development have been evaluated for method robustness under different sampling conditions. Differences in percent label dissolved between single-point sampling and profile sampling were observed for other methods, and investigations were performed to minimize the problems. The results of this investigation have influenced how we conduct dissolution method development. ACKNOWLEDGMENT The authors wish to acknowledge the insightful discussions provided by Vishwas Nesarikar, Robert Perrone, Scott Jennings, Ruben Lozano, Thomas Raglione, Pankaj Shah, Shufang Niu, and other members of our research groups here at Bristol-Myers Squibb. REFERENCES 1. Underwood, F.; Cadwallder, D. Effects of various hydrodynamic conditions on dissolution rate determination. J. Pharm. Sci. 1976, 65, 697. 2. Baxter, J. L.; Kukura, J.; Muzzio, F. J. Hydrodynamic-induced variability in the USP Apparatus II dissolution test. Int. J. Pharm. 2005, 292, 17 28. 3. Wu, Y.; Kildsig, D. O.; Ghaly, E. S. Effect of hydrodynamic environment on tablets dissolution rate. Pharm. Dev. Technol. 2004, 9 (1), 25 37. 4. McCarthy, L. G.; Bradley, G.; Sexton, J. C.; Corrigan, O. I.; Healy, A. M. Computational fluid dynamic modeling of the paddle dissolution apparatus: agitation rate, mixing patterns, and fluid velocities. AAPS PharmSciTech 2004, 5 (2), Article 31. 5. D Arcy, D. M.; Corrigan, O. I.; Healy, A. M. Hydrodynamic simulation of asymmetrically positioned tablets in the paddle dissolution apparatus: impact on dissolution rate and variability. J. Pharm. Pharmacol. 2005, 57 (10), 1243 1250. 6. Healy, A. M.; McCarthy, L. G.; Gallagher, K. M.; Corrigan, O. I. Sensitivity of dissolution rate to location in the paddle dissolution apparatus. J. Pharm. Pharmacol. 2002, 54, 441. 7. Lu, X.; Lozano, R.; Shah, P. A. In situ dissolution testing using different UV fiber optic probes and instruments. Dissolution Technol. 2003, 10 (4), 6 15. 8. Savage, T. S.; Wells, C. E. Automated sampling of in vitro dissolution medium: effect of sampling probes on dissolution rate of prednisone tablets. J. Pharm. Sci. 1982, 71 (6), 670 673. 9. Shah, V. P.; Gurbarg, M.; Noory, A.; Dighe, S.; Skelly, J. P. Influence of higher rates of agitation on release patterns of immediate-release drug products. J. Pharm. Sci. 1992, 81 (6), 500 503. 10. Lozano, R.; Joseph, J. M.; Kline, B. J. Temperature, ph and agitation rate as dissolution test discriminators of zofenopril calcium tablets. J. Pharm. Biomed. Anal. 1994, 12 (2), 173 177. 31 diss-14-04-08.indd 31 11/8/2007 1:57:50 PM