A new definition of pharmaceutical quality: Assembly of a risk simulation platform to investigate the impact of manufacturing/product variability on clinical performance (original) (raw)
Abstract
The absence of a unanimous, industry-specific definition of quality is, to a certain degree, impeding the progress of ongoing efforts to ''modernize'' the pharmaceutical industry. This work was predicated on requests by Dr. Woodcock (FDA) to re-define pharmaceutical quality in terms of risk by linking production characteristics to clinical attributes. A risk simulation platform that integrates population statistics, drug delivery system characteristics, dosing guidelines, patient compliance estimates, production metrics, and pharmacokinetic, pharmacodynamic, and in vitro-in vivo correlation models to investigate the impact of manufacturing variability on clinical performance of a model extended-release theophylline solid oral dosage system was developed. Manufacturing was characterized by inter-and intra-batch content uniformity and dissolution variability metrics, while clinical performance was described by a probabilistic pharmacodynamic model that expressed the probability of inefficacy and toxicity as a function of plasma concentrations. Least-squares regression revealed that both patient compliance variables, percent of doses taken and dosing time variability, significantly impacted efficacy and toxicity. Additionally, intra-batch content uniformity variability elicited a significant change in risk scores for the two adverse events and, therefore, was identified as a critical quality attribute. The proposed methodology demonstrates that pharmaceutical quality can be recast to explicitly reflect clinical performance. ß
Figures (10)
Figure 1. Schematic of the various model components that comprise the risk simulation platform. Figure adapted from Cogdill and Drennen.* Solid arrows represent compo- nents that are currently linked in the platform, whereas dotted arrows signify components/concepts that have yet to be incorporated.
Figure 2. Clearance cascade detailing the average theophylline clearance for indivi- duals classified according to numerous factors. Figure was adapted from Jusko et al.® Both the number of individuals in the original study by Jusko et al. and the percentage of the 100,000 simulated population that fell within each node are indicated. All terminal nodes are shaded. 0, 1, and 2 signifies the extensiveness of a given factor as delineated in the original study. MJ, Marijuana; OC, oral contraceptive; EtOH, alcohol; CHF, con- gestive heart failure; CIG, cigarette smoker; BENZ, benzodiazepines; BARBS, barbi- turates. A solid oral theophylline dosage system that was previously formulated and processed at Duquesne University (Pittsburgh, PA) and compacted at a local Once all of the factors were accounted for, theophyl- line clearance was individualized for each patient according to the clearance cascade adapted from Jusko et al.® (Fig. 2). The terminal node on the clearance cascade was determined for each patient based on the individualized factors that predispose theophylline disposition. The percentage of the total 100,000-patient population that fell within each node is reported in Figure 2. Once it was determined which node best described a given patient, the mean and standard deviation of that particular node (Fig. 2) were used to generate a normal distribution, from which a single value, representing the individual patient’s theophylline clearance, was _ extracted. Clearance estimates were restricted to 5-180 mL/h/
Figure 3. Frequency histograms of clearance (a) and volume of distribution (b) for the 100,000 simulated patients. The mean, standard deviation (SD), and range [ , ] of each parameter are also provided.
Analogous to the work of Buchwald,”° theophylline input was modeled using sigmoidal lag time and cut- off coefficients where absorption was assumed to be 100% of the maximum rate after 0.5h (i.e., lag time) and the absorption potential was reduced to 50% after 8h (ie., cut-off) to simulated time-dependent phe- nomena. These coefficients were used to adjust the input (J) of theophylline through the following series of equations Eq. (4d) is analogous to the original PK model (Eq. 4) with the exception of the lag time and cut-off terms. A numerical solution to | simulation time point ential equation solver. iq. (4d) was obtained for each using a Matlab-based differ-
Table 1. Summary of the Manufacturing Variability Metrics and the Treatment Parameters Used During Simulation
Figure 4. Plots of inefficacy (a) and toxicity (b) risk scores versus the fraction of observations for the sample population tested. These data were interpolated (solid lines) to determine the 95th percentile for inefficacy and toxicity.
where RS indicates the risk score for the ith observation and n represents the number of patients assessed. Patients were consecutively tested until the varia- bility of the risk estimates for both inefficacy and toxicity were below the threshold of 10°*. Further- more, the absolute change was required to retain a value below the threshold for 250 consecutive patients before the simulator converged on the risk estimates; these criteria were required for both inefficacy and toxicity. By this method, two risk scores, one for inefficacy and one for toxicity, were generated for each simulation trial.
Figure 5. Plots of inefficacy (a) and toxicity (b) risk scores versus various dissolution time constants tested in different age-restricted sample populations.
“Metric—mean (%): 25.96; SD (%): 0.24; SEM (%): 0.017; upper 95%, mean (%): 26.00; lower 95%, mean (%): 25.93; number of observations: 192. >Metric—mean (%): 7.43; SD (%): 0.39; SEM (%): 0.028; upper 95%, mean (%): 7.48; lower 95%, mean (%): 7.87; number of observations: 192. Table 2. Summary Statistics for the 6-Factor Full Factorial Experimental Design
Figure 6. Plots of the predicted mean probabilities for inefficacy (a and b) and toxicity (c and d) adjusted for the effects of intra-batch content uniformity variability, patient compliance, and dosing time standard deviation. Asterisks denote the upper and lower values of the mean confidence intervals whereas the open circles represent the mid-point of the intervals.
Loading Preview
Sorry, preview is currently unavailable. You can download the paper by clicking the button above.
References (31)
- Cogdill RP, Knight TP, Anderson CA, Drennen JK III. 2007. The financial returns of investments in process analytical technology and lean manufacturing: Benchmarks and case study. J Pharm Innov 2:38-50.
- 2004. Pharmaceutical CGMPs for the 21st Century-A risk- based approach final report. Rockville: U.S. Department of Health and Human Services, Food and Drug Administration.
- Woodcock J. 2004. The concept of pharmaceutical quality. Am Pharm Rev 7:10-15.
- Cogdill RP, Drennen JK. 2008. Risk-based quality by design (QbD): A Taguchi perspective on the assessment of product quality, and the quantitative linkage of drug product para- meters and clinical performance. J Pharm Innov 3:23-29.
- Bonate PL. 2001. A brief introduction to Monte Carlo simula- tion. Clin Pharmacokinet 40:15-22.
- Ahmad AM, Boudinot FD, Barr WH, Reed RC, Garnett WR. 2005. The use of Monte Carlo simulations to study the effect of poor compliance on the steady state concentrations of Valproic Acid following administration of enteric-coated and extended release Divalproex Sodium formulations. Biopharm Drug Dis- pos 26:417-425.
- Short SM. 2009. Performance-based quality specifications: The link between product development and clinical outcomes. PhD Dissertation. Pharmaceutical Sciences, Pittsburgh: Duquesne University. p 322.
- Jusko WJ, Gardner MJ, Mangione A, Schentag JJ, Koup JR, Vance JW. 1979. Factors affecting theophylline clearances: Age, tobacco, marijuana, cirrhosis, congestive heart failure, obesity, oral contraceptives, benzodiazepines, barbiturates, and ethanol. J Pharm Sci 68:1358-1366.
- Gilman TM, Muir KT, Jung RC, Walberg CB. 1985. Estimation of theophylline clearance during intravenous aminophylline infusions. J Pharm Sci 74:508-514.
- Powell JR, Vozeh S, Hopewell P, Costello J, Sheiner LB, Riegel- man S. 1978. Theophylline disposition in acutely ill hospitalized patients. The effect of smoking, heart failure, severe airway obstruction, and pneumonia. Am Rev Respir Dis 118:229- 238.
- Voytilla RJ. 2007. Non-destructive prediction of drug release from tablets using near-infrared spectroscopy. MS Thesis. Graduate School of Pharmaceutical Sciences, Pittsburgh: Duquesne University. p 212.
- Umesh B. 1992. Pharmaceutical dissolution testing. New York: Marcel Dekker. p 437.
- Hussein Z, Friedman M. 1990. Release and absorption char- acteristics of novel theophylline sustained-release formula- tions: In vitro-in vivo correlation. Pharm Res 7:1167- 1171.
- Yamazaki M, Fukutomi O, Kondo N, Kato Z, Nakashima Y, Shinoda S, Agata H, Kondo T, Imaeda N, Orii T. 1994. The design of oral sustained-release theophylline dosing after con- version from intravenous to oral therapy. Int J Clin Pharmacol Ther 32:625-631.
- Weinberger MM. 1984. Theophylline QID, TID, BID and now QD? A report on 24-hour dosing with slow-release theophylline formulations with emphasis on analyses of data used to obtain Food and Drug Administration approval for Theo-24. Pharma- cotherapy 4:181-198.
- Sabnis S, Adeyeye CM. 1998. Controlled-release hydrophilic tablets for individualized theophylline therapy. Drug Dev Ind Pharm 25:187-196.
- Ohnishi A, Kato M, Kojima J, Ushiama H, Yoneko M, Kawai H. 2003. Differential pharmacokinetics of theophylline in elderly patients. Drugs Aging 20:71-84.
- Mitenko PA, Ogilvie RI. 1973. Rational intravenous doses of theophylline. N Engl J Med 289:600-603.
- Weinberger M, Hendeles L, Bighley L. 1978. The relation of product formulation to absorption of oral Theophylline. N Engl J Med 299:852-857.
- Takayama K, Morva A, Fujikawa M, Hattori Y, Obata Y, Nagai T. 2000. Formula optimization of theophylline controlled- release tablet based on artificial neural networks. J Control Release 68:175-186.
- Chiou WL, Gadalla MAF, Peng GW. 1978. Method for the rapid estimation of the total body drug clearance and adjustment of dosage regimens in patients during a con- stant-rate intravenous infusion. J Pharmacokinet Bio- pharm 6:135-151.
- Shargel L, Yu ABC. 1999. Applied biopharmaceutics and phar- macokinetics, 4th edition. Stamford: Appleton & Lange. p 768.
- Buchwald P. 2003. Direct, differential-equation-based in-vitro- in-vivo correlation (IVIVC) method. J Pharm Pharmacol 55:495-504.
- DiPiro JT, Spruill WJ, Blouin RA, Pruemer JM. 2002. Concepts in clinical pharmacokinetics, 3rd edition. Bethesda: American Society of Health-System Pharmacists, Inc. p 279.
- 2008. Pharmaceutical Development Q8(R1). ICH Harmonised Tripartite Guideline.
- 2004. Innovation/stagnation: Challenge and opportunity on the critical path to new medical products. Rockville: U.S. Depart- ment of Health and Human Services, Food and Drug Admin- istration.
- 2003. Comparability protocols-Protein drug products and biological products-Chemistry, manufacturing, and con- trols information DRAFT GUIDANCE. Rockville: U.S. Department of Health and Human Services, Food and Drug Administration, Center for Biologics Evaluation and Research, Center for Drug Evaluation and Research, Center for Veterinary Medicine.
- Bonate PL. 2000. Clinical trial simulation in drug development. Pharm Res 17:252-256.
- Grass GM, Sinko PJ. 2002. Physiologically-based pharmacoki- netic simulation modelling. Adv Drug Deliv Rev 54:433-451.
- Holford NHG, Kimko HC, Monteleone JPR, Peck CC. 2000. Simulation of clinical trials. Annu Rev Pharmacol Toxicol 40:209-234.
- Poland B, Wada R. 2001. Combining drug-disease and economic modelling to inform drug development decisions. Drug Discov Today 6:1165-1170.