Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

J Pharm Innov (2008) 3:23–29

DOI 10.1007/s12247-008-9025-3

PERSPECTIVE

Risk-based Quality by Design (QbD): A Taguchi Perspective


on the Assessment of Product Quality, and the Quantitative
Linkage of Drug Product Parameters and Clinical
Performance
Robert P. Cogdill & James K. Drennen

Published online: 6 March 2008


# International Society for Pharmaceutical Engineering 2008

Abstract For good reason, quality by design (QbD) has guidance [1] as “building in quality from the development
become a topic of significant interest within the pharmaceu- phase and throughout a product’s life cycle.” According to
tical industry. Whereas regulatory agencies and standard- the same guidance, (implementing) QbD means “designing
setting organizations are moving swiftly to establish QbD and developing a product and associated manufacturing
guidance relevant to the needs of pharmaceutical manufac- processes that will be used during product development to
turing, much of the core science of QbD was established long ensure that the product consistently attains a predefined
ago by Dr. Genichi Taguchi and other leaders in the field of quality at the end of the manufacturing process.” Another
quality management. In order for the pharmaceutical definition of QbD is given by FDA’s report on Critical Path
industry to fully capitalize on QbD, new methods for Opportunities for Generic Drugs, which states that “under
quantitatively assessing product quality need to be estab- the QbD paradigm, quality is built into the final product by
lished. This perspective article presents a risk-based strategy understanding and controlling formulation and manufactur-
for quality assessment which uses model-based simulation to ing variables: testing is used to confirm the quality of the
link variation in drug product parameters and clinical product” [2]. The recent revision (R1) of the ICH Q8
performance. This work is intended to provide background guideline defines QbD as a systematic approach to
and introduction for a series of manuscripts dealing with development that begins with predefined objectives, and
QbD based on probabilistic risk assessment. emphasizes product and process understanding and process
control, based on sound science and quality risk manage-
Keywords Quality by design (QbD) . ment [3]. The differences between the current and desired
Taguchi methods . Design space . PAT . Specifications . states of pharmaceutical manufacturing (regarding QbD)
Pharmaceutical development . Quality loss function (QLF) are summarized in [3].
Incorporating QbD into development activities is strate-
gically important because it is the prerequisite for the
Introduction establishment of an operating design space, and it is
perhaps the most direct means of communicating process
Quality by Design understanding. The establishment of an extensive design
space can be an important driver of efficiency because an
Quality by design (QbD), which has become a topic of operational adjustment within the (approved) design space
significant interest within the pharmaceutical industry, is is not considered a change; therefore, reducing the need to
described by Food and Drug Administration (FDA) submit a post approval supplemental filing. Furthermore,
the capability to operate flexibly within an established
design space is essential to achieving true continuous
R. P. Cogdill : J. K. Drennen (*)
improvement [4–6].
Duquesne University Center for Pharmaceutical Technology,
Pittsburgh, PA, USA The pharmaceutical industry has moved very quickly
e-mail: drennen@duq.edu to begin working to incorporate QbD into development
24 J Pharm Innov (2008) 3:23–29

protocols and, in some respects, is accelerating regulatory reiterated [10]) that the lack of observable connection
changes because of the demand for guidance. For example, between critical product attributes and clinical performance
the 2005 Step 4 version of the International Conference on is a persistent roadblock to establishing an effective, risk-
Harmonization (ICH) Q8 Guideline on Pharmaceutical based definition of pharmaceutical quality. The objectives
Development [7], which is often cited in reference to of this manuscript are to (1) describe the connections
QbD by FDA guidance, does not specifically mention QbD. between Taguchi’s concepts of quality loss, pharmaceutical
Rather, the guideline states that “it is important to recognize quality, and risk, and (2) propose a simulation framework
that quality cannot be tested into products; i.e., quality for the assessment of pharmaceutical quality based on
should be built in by design.” This is substantially probabilistic risk assessment.
addressed by the R1 revision of the Q8 Guideline, however,
which provides significant clarification of aspects related to Taguchi Methods and QbD
QbD, including harmonized definitions for critical quality
attributes, critical process parameters, real-time release, and Quality by design is not a new concept [11–16]. Indeed, Dr.
methods which can be used to define a design space [3]. Genichi Taguchi, who has been credited by some as the
In 2005, FDA began a pilot program which enables father of QbD, began using designed experiments based on
participating firms to submit chemistry, manufacturing, orthogonal arrays while working as a consultant for
and controls (CMC) information demonstrating applica- Morinaga Pharmaceuticals Company of Japan from 1947
tion of QbD using the quality overall summary (QOS) to 1949. While at Morinaga, Taguchi was taught DOE and
format [8]. The CMC pilot project provides a mechanism statistical methods by Motosaburo Masuyama, who was
for FDA and industry to cooperate on strategies for the leading an effort to improve the quality and yield of the
implementation of the ICH Q8 and Q9 guidelines, and industrial production of penicillin.
FDA’s PAT guidance. According to a recent interview with Whereas Dr. Taguchi’s earliest professional work was
Dr. Chi-wan Chen [8], deputy director of the Office of New concerned with statistical sampling theory [11], his greatest
Drug Quality Assessment (ONDQA), considerable diversity contributions to industrial technology are associated with
was observed in the way QbD and the concept of design his lifelong work to advance the science of robust design.
space is approached by industry participants. The Japanese quality revolution began with the prolifera-
Chen described FDA’s view of QbD as a “systematic tion of Dr. Walter Shewhart’s techniques for statistical
approach to product and process design and development” process control (SPC), which were taught to Japanese
involving seven key elements: engineers and scientists beginning in the late 1940s by a
Shewhart disciple, W. Edward Deming [16]. Taguchi recog-
& Identification of target product profile
nized that, whereas online methods of quality improvement,
& Determination of product critical quality attributes (CQAs)
such as SPC, are highly effective in bringing existing
& Linkage of raw material attributes and process param-
processes into control, thereby reducing waste and the need
eters to CQAs
for end-of-process inspection, the most efficient point
& Risk assessment
during the product lifecycle to achieve high quality is not
& Design space
during production, but during design.
& Design and implementation of a control strategy
Taguchi defines the development of a product or process
& Product lifecycle management and continuous improvement
as a three-stage operation: system design, parameter design,
Whereas the main elements and specific techniques of and tolerance design [12, 13]. In relation to pharmaceutical
QbD, such as design of experiments (DOE) or failure mode development, system design includes, for example, select-
effect analysis (FMEA), have been described in detail, ing the active ingredient and drug delivery platform to
much less discussion has been devoted on how to define the achieve a specific pharmacodynamic target (defined by
target product profile, which is perhaps the most unique and efficacy and lack of toxicity). The concept of system design
challenging aspect of QbD applied to pharmaceutical is conveyed in the ICH Q8 guideline for pharmaceutical
manufacturing. In other words, before QbD can be achieved development, which defines quality as “the suitability of
for a particular product or process, the true meaning of what either a drug substance or drug product for its intended use”
defines quality, i.e., the voice of the consumer, must be [7]. Effective system design begins with a quantitative
known. understanding of target quality, followed by identification
In a 2004 perspective article [9], Janet Woodcock of a product or process capable of reliably achieving the
described the problems and limitations of the current target.
definition (or lack thereof) of pharmaceutical quality and Tolerance design corresponds to setting the specifica-
suggested that the concept of pharmaceutical quality should tions for raw and in-process materials necessary to achieve
be recast in terms of risk. She observed (and recently finished product quality targets. When variations in certain
J Pharm Innov (2008) 3:23–29 25

raw material attributes are observed to have a critical effect fications. Process capability, which is currently used as a
on finished product quality or when the true range or effect target metric for process improvement and benchmarking
of variability is unknown, the typical engineering response studies, is based on the concept of conformance with
is to tighten the corresponding raw material acceptance specifications. The problem with pass/fail specifications, as
specifications. Taguchi observed, is that no distinction is made between
Taguchi’s approach to development is unique, however, the quality of an item produced exactly on target and an
because of his focus on parameter design. During parameter item which just meets specifications (Figs. 1 and 2).
design, the engineer seeks to optimize the system design Taguchi defines the quality of a product as “the (minimum)
through simple, offline experiments to identify configura- loss imparted by the product to the society from the time
tions which minimize performance variation in the presence the product is shipped” [11]. Accordingly, any product
of uncontrollable variance factors or noise. The objective of deviation from the target design results in greater loss and,
parameter design is to reduce the need to rely on restrictive hence, lower quality. Taguchi sought to incorporate not
tolerance specifications to achieve quality, which is the only direct costs, such as material waste, but also indirect
essence of ‘building quality into’ a design. Taguchi adapted costs, such as restocking fees, loss of brand image, or losses
concepts from fields such as statistics and signal processing experienced by the customer. Mathematically, Taguchi’s
to assemble a toolbox of simple techniques an engineer QLF is essentially a calculation of the mean squared
could use to quickly perform parameter design using deviation or error (MSE) between observed and target
limited resources. Taguchi’s methods include linear graphs, attribute values. Whereas Taguchi’s QLF is admittedly
orthogonal arrays, and the signal-to-noise (S/N) ratio for the simplistic, some empirical studies have demonstrated that
identification of the control parameters. the formula is often accurate within reasonable limits [11].
The continuous QLF is an important metric for design
Taguchi’s Continuous Quality Loss Function because it provides the engineer with a simple formula for
estimating the cost of variability and, hence, a means of
An essential component of Taguchi’s methods is his determining the value of investments in quality improve-
concept of the continuous quality loss function (QLF), ment. Taguchi intended that practitioners would ‘calibrate’
which provides the numerical basis for his S/N technique. the QLF for a particular product or process by estimating
Before the widespread use of Taguchi’s methods within the actual loss related to one or more levels of deviation,
Japanese industry, the prevailing quality assessment method thereby transforming units of product attribute variability
was pass/fail testing to determine conformance with speci- into a single measurement system based on cost. For many

Fig. 1 Graphical comparison of pass/fail specifications and (Taguchi’s) systems based on Taguchi’s QLF only achieve minimum quality loss
continuous QLF. Whereas systems based on pass/fail specification testing when the target value is achieved. In a risk-based system, the shape of the
do not distinguish between quality levels within acceptance limits, QLF could be determined by risk assessment simulation
26 J Pharm Innov (2008) 3:23–29

however, the markets for pharmaceutical drugs (and health-


care in general) are not efficient.
Patients are generally unable to directly determine the
quality of an individual drug product before consumption
and often assume that quality is uniform (i.e., assured by
industrial and regulatory oversight) [9]. Consider, for
example, the persistent willingness of consumers to
purchase prescription drugs via the Internet, which offers
scant assurance of product condition or pedigree [17]. In
addition, purchase decisions are often driven by external
factors, such as physician recommendation, insurance
coverage, or lack of alternative treatments rather than
perception of superior quality. In other words, the con-
sumer’s response to quality is inefficient relative to the
manufacturer’s response to production cost. Therefore, the
economic value of quality is less determined by consumer
demand than by regulatory oversight for which product cost
has not always been a concern (as costs are borne by the
manufacturer, consumer, and society, not the regulator).
Fig. 2 Schematic representation of the limitations of measuring Rather than economic cost, risk provides a more
product quality using multiple criteria without considering their
appropriate basis for the measurement and valuation of
interaction. Whereas both tablets A and B are very close to product
quality acceptance limits (perhaps within the limit of measurement drug product quality as manufacturers, patients, and
precision), only tablet A would be graded as acceptable, despite the regulators are uniformly aligned in their desire to minimize
fact that it likely poses greater pharmaceutical risk than tablet B the sources of risk associated with drug therapy (Fig. 3). At
the most basic level, patients seek safe, effective, and
affordable drug therapy; all other considerations (e.g., taste
products, the use of economic loss as a basis for quality or appearance) are essentially subordinate. Within a risk-
measurement facilitates accurate prioritization of variance based quality measurement system, the mean and variance
factors and financial optimization of both product and of product attributes, such as API content and the rate of
process without sacrificing consideration of quality. drug release, are transformed into estimates of risk because
of toxicity (e.g., superpotency) and inefficacy (e.g., incom-
Risk as a Basis for Measurement of Drug Quality plete release). Therefore, the objectives of product, process,
and specification design would uniformly be to minimize
Whereas the concept of a continuous QLF is important for risk.
the implementation of QbD during drug development, An advantage of risk-based quality measurement is that
financial cost is not an appropriate basis for the measure- it provides an ethical way of weighing the value of
ment of quality loss associated with pharmaceutical additional quality assurance expenditures. Consider that,
products. In an efficient market, the point of optimal whereas we have assumed nearly inelastic demand sensi-
economic operation is where the marginal cost and tivity to quality variation, many healthcare economists have
marginal benefit of incremental investment in quality demonstrated that patients do exhibit price elasticity in their
improvement are equal. In the traditional economic sense, purchase decisions, even when a third party payer (e.g.,

Fig. 3 Sources of risk from


medicine. FDA-CDER 2005
Report to the Nation
J Pharm Innov (2008) 3:23–29 27

more, risk related to product cost would only impact design


decisions once additional investment in quality assurance
no longer provided meaningful risk reduction. Taguchi,
who was notoriously cost-conscious, is noted as saying that
“it’s just as unethical to add tremendous cost to ensure
products are of good quality as it is to ship defective goods”
[11]. Furthermore, in addition to protecting public health by
assuring the safety, efficacy, and security of drug products,
the FDA is (also) responsible for advancing public health
by helping to speed innovations that make medicines more
effective, safer, and more affordable [21]. As stated at the
end of FDA’s 2004 Critical Path Initiative report, (industry,
academia, and FDA) must work together to find…less
costly ways to turn good biomedical ideas into safe and
Fig. 4 Schematic view of the models and their connections as part of
a Monte Carlo probabilistic risk assessment system effective treatments, (otherwise) “the hoped-for benefits of
the biomedical century may not come to pass, or may not
be affordable” [21]. Finally, when asked about the critical
prescription insurance) is involved [18–20]. Thus, if the success factors for the industry and regulatory agencies
price elasticity of demand for a given product can be over the next 5 to 10 years, Dr. Moheb Nasr, Director of the
estimated, incremental price increases might be transformed Office of New Drug Quality Assessment, stated that the
into a measure of risk analogous to the risk of inefficacy (or “ultimate success (for industry and regulators) is having
lack of availability). This would create an ethical incentive affordable medicines for the patient” [4].
to optimize the alignment of regulatory and financial
priorities. Simulation for Quality Risk Assessment
Whereas the relationship of cost to risk is obviously
indirect, the concept of indirect risk is similar to Taguchi’s Consider the potential risk consequences related to the
use of indirect costs in determining quality loss. Further- specification of product quality acceptance criteria. Insuffi-

Fig. 5 Example of the effect of product quality variability (content achieving steady state, for example, or the average deviation from
uniformity) on chronic dosing of a model drug (theophylline) target concentration. Process sigma, σ, is defined such that 3σ is equal
generated by Monte Carlo simulation. Risk might be measured as to 1.0 Cpk or 2,700 defects per million opportunities (DPMO)
the probability of excursion out of the therapeutic range after
28 J Pharm Innov (2008) 3:23–29

and effectiveness of treatment. Finally, the limitations (lag


time, sampling, precision) of systems for measuring both
product characteristics and patient response can mask subtle
(yet relevant) correlations. These and other limiting factors
have been described previously by Woodcock [9].
Mathematical models form the language used to com-
municate the connections between fundamentals and
observed phenomena. Models are advantageous because
they allow greater understanding of processes and systems,
yet provide users a platform to easily manipulate the inputs
to examine the outputs [22]. To the extent that a fun-
damentally relevant model can be identified, model
coefficients provide an efficient means of separating signal
from noise. Model-based representations of patient, phar-
maceutical, pharmacokinetic (PK), and pharmacodynamic
(PD) systems are the key to overcoming the finite limits of
sampling when trying to assess the clinical relevance of
quality. Models have long been used successfully to
describe the dynamics of drug release, absorption, distribu-
tion, metabolism, and elimination (ADME), and patient
Fig. 6 Example of how a risk-based system of product quality
assessment might be implemented. Depending on the nature of the response.
product (and how it will be prescribed) the region of acceptable risk Monte Carlo simulation is a stochastic modeling tech-
might be similar to or significantly more or less extensive than an nique often used for probabilistic risk assessment, which
acceptance region based on (univariate) specifications offers a flexible mathematical framework to harness the
various models (and probability density functions) neces-
cient quantitative understanding of how product variability sary to link product quality and clinical risk. Monte Carlo
affects the safety and efficacy risks of treatment can result in simulation differs from traditional simulation in that the
the specification of product quality acceptance criteria which model parameters are treated as stochastic or random
do not sufficiently mitigate risks to patient harm; in such a variables, rather than fixed values [22]. Figure 4 illustrates
case, a risk-based quality design system should indicate how the various models representing drug products and
that a net risk reduction could be achieved by tightening
specifications. Alternatively, however, it is plausible that
insufficient understanding more frequently leads to the
imposition of excessively strict tolerances as a way of
mitigating the unknown level of risk [9, 21]. Comprehensive
risk analysis would show that the amount of risk mitigated
by unrealistically restrictive design tolerances (or standards)
does not offset the negative effects on patients because of
reduced affordability (or availability) of treatment.
Whereas appropriate dosing guidelines are routinely
determined by carefully balancing the relationship between
dose, efficacy, and toxicity for a given population of
patients, it is considerably more difficult to determine the
quality cost imposed by variation in drug product attributes.
First, the criticality of variation in factors, such as API
content and rate of drug release, are interrelated and often
patient-specific. For example, the probability of ineffective
treatment posed by a subpotent tablet will be magnified if Fig. 7 Example of how product quality variability, measured as
the rate or extent of drug release is low and will be further process sigma, might be correlated to clinical performance. The
exacerbated if the patient’s rate of drug elimination is high. synthetic data used for this example did not include variability in
factors other than drug content uniformity. Because there are 2 df in
Second, the dynamics of repeated dosing and patient
process capability measurement and, hence, some ambiguity in the
compliance complicate the relationship between variation data, clinical risk based on simulation may be a more effective
in the attributes of individual doses and the overall safety measure of quality to guide operational or design changes
J Pharm Innov (2008) 3:23–29 29

patients could be linked for the purpose of simulating the MD: U.S. Department of Health and Human Services, Food and Drug
clinical effect of changes in drug product design or quality. Administration; 2007.
3. Q8 (R1): Pharmaceutical Development, Revision 1. ICH Harmonized
If repeated dosing is assumed, for example, the pharma- Tripartite Guidelines. International Conference on Harmonization of
cokinetic concept of superposition could be used to Technical Requirements for Registration of Pharmaceuticals for
estimate the relationship between content uniformity and Human Use; 2007.
the probability of exceeding the therapeutic window of 4. Hall G, Runas R. JPI Interviews Moheb Nasr, PhD. J Pharm
Innov. 2007;2(3–4):67–70.
target plasma drug concentration (Fig. 5). The model-based 5. U.S. Department of Health and Human Services, Food and Drug
system could, therefore, be used to set “realistic” target Administration. Pharmaceutical CGMPs for the 21st century—a risk-
values for drug quality attributes (Fig. 6) or manufacturing based approach, final report. Rockville, MD: U.S. Department of
performance (Fig. 7). Ultimately, the goals of risk modeling Health and Human Services, Food and Drug Administration; 2004.
6. U.S. Department of Health and Human Services, Food and Drug
would be to (1) optimize the product and process design for Administration. Guidance for industry: PAT—a framework for
minimum risk, (2) develop for each product a system for innovative pharmaceutical development, manufacturing, and
optimizing patient-specific dosing guidelines, and (3) quality assurance. Rockville, MD: U.S. Department of Health
generate a risk-based QLF or design space which could and Human Services, Food and Drug Administration; 2004.
7. Q8: Pharmaceutical Development. ICH Harmonized Tripartite Guide-
be paired with PAT and process control systems so that lines. International Conference on Harmonization of Technical Require-
production could be maintained in a low risk state. ments for Registration of Pharmaceuticals for Human Use; 2005.
8. Van Arnum P. A FDA perspective on quality by design. New York,
NY: PharmTech; 2007.
9. Woodcock J. The concept of pharmaceutical quality. Am Pharm
Summary Rev. 2004;7(6):10–15.
10. Woodcock J. International Federation of Process Analytical
Quality by design is an important current topic in Chemistry (IFPAC) Annual Meeting, Baltimore, MD, 28 January
pharmaceutical manufacturing because it is the logical next 2007.
11. Ealey LA. Quality by design: Taguchi methods and U.S. industry.
step in the path toward the desired state that began with the Dearborn, MI: ASI; 1988.
FDA’s PAT guidance and twenty-first century cGMPs 12. Ross PJ. Taguchi techniques for quality engineering. New York,
initiative. As was the case with PAT, the pharmaceutical NY: McGraw-Hill; 1988.
industry benefits from many years of pioneering work in 13. Taguchi G, Chowdhury S, Wu Y. Taguchi’s quality engineering
handbook. Hoboken, NJ: Wiley; 2005.
QbD by Taguchi and others. As has also been observed 14. Wu H, Khan MA, Hussain AS. Process control perspective for
with the introduction of PAT, however, the unique aspects process analytical technology: integration of chemical engineering
of pharmaceutical product design and manufacturing are practice into semiconductor and pharmaceutical industries. Chem
leading the industry to push the development of new Eng Commun. 2007;194:760–79.
15. Barker TB. Quality by experimental design. New York: Marcel
concepts and strategies for QbD. Dekker; 1985.
The objectives of this work were to (1) describe the 16. Marash SA, Berman P, Flynn M. Fusion management: harnessing
connections between Taguchi’s concepts of quality loss, the power of six sigma, Lean, ISO 9001:2000, Malcom Baldridge,
pharmaceutical quality, and risk, and (2) propose a TQM and other quality breakthroughs of the past century. Fairfax,
VA: QSU; 2004.
simulation framework for the assessment of pharmaceutical 17. U.S. Department of Health and Human Services, Food and Drug
quality based on probabilistic risk assessment. This manu- Administration. FDA says consumers continue to buy risky drugs
script is intended to precede a series of future articles which online. Rockville, MD: U.S. Department of Health and Human
will provide a more detailed demonstration of how risk- Services, Food and Drug Administration. http://www.fda.gov/bbs/
topics/NEWS/2007/NEW01735.html. November 1, 2007.
based QbD can be achieved and integrated with PAT 18. Domino ME, Salkever DS. Price elasticity and pharmaceutical
systems to enhance the development, production, and selection: the influence of managed care. Health Econ. 2003;12
quality assessment of pharmaceutical products. (7):565–86.
19. Gemmill MC, Costa-Font J, McGuire A. In search of a corrected
prescription drug elasticity estimate: a meta-regression approach.
Health Econ. 2007;16(6):627–643.
20. Vernon JA, Hughen WK, Trujillo AJ. Pharmaceutical manufac-
References turing efficiency, drug prices, and public health: examining the
causal links. Drug Inf J. 2007;41(2):229–39.
1. U.S. Department of Health and Human Services, Food and Drug 21. U.S. Department of Health and Human Services, Food and Drug
Administration. Guidance for industry: quality systems approach Administration. Challenge and opportunity on the critical path to
to pharmaceutical CGMP regulations. Rockville, MD: U.S. new medical products. Rockville, MD: U.S. Department of Health
Department of Health and Human Services, Food and Drug and Human Services, Food and Drug Administration. http://www.
Administration; 2006. fda.gov/oc/initiatives/criticalpath/whitepaper.pdf. March 2004.
2. U.S. Department of Health and Human Services, Food and Drug 22. Bonate PL. A brief introduction to Monte Carlo simulation. Clin
Administration. Critical path opportunities for generic drugs. Rockville, Pharmacokinet. 2001;40(1):15–22.

You might also like