You are on page 1of 8

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/269573886

Quality by Design (QbD) Approach in Pharmaceuticals: Status, Challenges and


Next Steps

Article  in  Current Drug Delivery · December 2014


DOI: 10.2174/2210303104666141112220253

CITATIONS READS
6 8,888

2 authors:

DR. Lalit Singh Dr Vijay Sharma


SRMS College of Engineering and Technology (Pharmacy) IFTM University
58 PUBLICATIONS   113 CITATIONS    46 PUBLICATIONS   144 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Optimization, Controlled Release, Gastroretentive Drug Delivery Systems, Hypoglycemic Drug(s) View project

Establishment and Escalation of Amino Acid Stacked Repressible Release Embedded System Using QbD View project

All content following this page was uploaded by Dr Vijay Sharma on 08 August 2015.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.ae
2 Drug Delivery Letters, 2015, 5, 2-8

Quality by Design (QbD) Approach in Pharmaceuticals: Status, Challenges


and Next Steps

Lalit Singh and Vijay Sharma*

Shri Ram Murti Smarak College of Engineering and Technology, Pharmacy, Bareilly, India

Abstract: Traditional approach of formulating a new drug product is an exhaustive task and involves a
number of resources like man, money, time and experimental efforts. While, using this Quality by
Design (QbD) approach one can get the pharmaceutical product of desired (best) quality with minimiz-
ing above resources as well as knowing the influence of one factor over the desired associated process.
Hence aim of this study is the understanding of QbD approach to design product and manufacturing
process to get desired pharmaceutical product. QbD follows the concepts of ICH guidelines (Q8, Q9 &
Q10) which are essential for processing a pharmaceutical process. This review emphasizes on various
aspects of keynotes of QbD like ascertaining drug product quality profile, priorizing input variables for
optimization, modelization & validation of QbD methodology and in the last QbD validation, scale up and production as
well as the software used for QbD. Hence, QbD approach is not only useful in facilitating comprehension of the products
or process but also results in an excellent and economical product which follows federal compliance.
Keywords: ICH guidelines, product quality profile, quality by design.

INTRODUCTION FDA report “Pharmaceutical cGMPs for the 21st Cen-


tury: A Risk-Based Approach” has made it imperative to use
FDA announced a new initiative in 2002 for risk man- quality by design (QbD) approach in pharmaceuticals [3]. In
agement i.e. (cGMP for the 21st Century: A Risk based Ap- present century, pharmaceutical industry is still using manu-
proach) to modernize the FDAs regulation for maintaining facturing technologies that have been employed since 1940s
better pharmaceutical quality as well as setting up new regu- which mostly resulted in inefficient, overly expensive proc-
latory framework focusing on QbD, risk management and esses that were ultimately not in the best long-term interests
quality maintaining system. QbD needs good knowledge of of patients. Even the industries did not make significant
final product and in-process or process variables that affect changes in manufacturing process unless the significant
end-product quality. This newer concept being considered by compliance or cost saving advantages could justify the high
FDA in its current-GMP initiative, two main documents costs and long cycle time that was needed to gain approval.
were generated by International Conference on Harmoniza- The quality may not be completely ensured with use of qual-
tion (ICH), to guide the quality i.e. ICH-Q8: (Pharmaceutical ity by testing (QbT) on finished product (Fig. 1) which was
Development) and ICH-Q9: (Quality Risk Management). used in early stage in good manufacturing practices (GMPs).
The pharmaceutical industry has been a highly regulated On the other hand, current good manufacturing practices
industry in the past for many good reasons [1]. The major (cGMP) acknowledged the undesired impact of GMPs on
hurdles in accomplishing quality in a product can be attrib- understanding manufacturing science and sought to set the
uted to the cumulative variability in drug substance(s), ex- framework for additional guidance that encouraged risk- and
cipients, process(es), packaging material(s), and so on, as science-based understanding. Thus, it allows more freedom
illustrated in (Fig. 1) (Sources of Variability). The pharma- to introduce innovations and improvements that will result in
ceuticals have greatly improved the mortality and morbidity enhanced quality, cost, or timing [4].
rates, but purity, potency, delivery rate, and other properties
A new paradigm for regulation and time to market new
of medicine have been mitigated by various elements of risk
and improved medicines, to describe the comprehensive dis-
to the patients. Pharmaceutical regulations have clearly pro-
cipline, are required to shut down the quality problem [5].
tected the population from much of the needless harm as
Juran proposed the concept of QbD which was first outlined
incurred early in the twentieth century; there has been a con-
in the 1960s. According to this concept, quality could be
cern more recently that overregulation may be associated
something that may be planned rather than simply achieved
with stifling innovation that can improve pharmaceutical in (more often than not) a haphazard way [6].
quality even further [2].
The concept mentioned in the ICH-Q8 guideline, states
that “quality of product cannot be tested”. Systematic ap-
*Address correspondence to this author at the Shri Ram Murti Smarak College proach of QbD begins with predefined objectives and under-
of Engineering and Technology, Pharmacy, Bareilly, India; Tel: 9458702561, standing of product and its process as well as process control
Fax: 0581-2582330; E-mail: vijaysrampur@gmail.com which was based on sound knowledge of science and quality

2210-303X/15 $58.00+.00 © 2015 Bentham Science Publishers


Quality by Design (QbD) Approach in Pharmaceuticals: Status, Challenges and Next Steps Drug Delivery Letters, 2015, Vol. 5, No. 1 3

risk management [7]. Formulation parameters (independent changed to get drug product of desired characteristics. For
variables) affect the product’s characteristics and are useful developing a quality drug product, and getting a relationship
in optimization of independent variables in order to monitor between formulations and their manufacturing process vari-
the behavior of dependent variables in producing the opti- ables including active pharmaceutical ingredients, excipients,
mized product, under the given set of conditions. Thus, vari- process variables, desired product characteristics and sources
ables are included in QbD to ease the final best formulation of variability. This knowledge is then used to develop a
of drug [8]. product of desired characteristics [10-13].
Although the idea of QbD is not new but it is still a sound
tool in the pharmaceutical industry, as it has capability to
profoundly change how drugs products are discovered, de-
veloped, manufactured and even regulated. Most crises or
failings around quality, as per Juran are simply related to
how processes were planned, set up, embedded and man-
aged. Knowledge management and quality risk management
are two of the primary enablers of QbD. They play a critical
role both in development and in the implementation of QbD.
They are instrumental in achieving final product, establish-
ing and maintaining a state of art for control, and lastly imi-
tating continual improvement [14].

BACKGROUND OF QUALITY BY DESIGN


ICH-Q8: (pharmaceutical development) is the guideline
to understand the concept of QbD. It defines QbD as “QbD
is a systematic approach to design a product of predefined
quality and its production process to continually and consis-
Fig. (1). Various sources of variability.
tently delivering intended performance of the final product.
The data collected from pharmaceutical development studies
Later in June 2007, Product Quality Lifecycle Implemen- and manufacturing experiences, utilized for logical under-
tation (PQLI) was laid down by the International Society for standing for design space its specifications and process con-
Pharmaceutical Engineering (ISPE), in a workshop, which trols, based on sound science and quality risk management”.
was held in Europe in September. The PQLI was imple- ICH-Q8:(R1) and ICH-Q8:(R2) guidance are the revision of
mented in US followed with the objective of coordinating ICH-Q8, and an annex to ICH-Q8. It provides further
with the various regulatory agencies throughout the world to clarification of key concepts outlined in the core guideline
encourage a common intellectual understanding of QbD. The and describes the principles of QBD.
aim of PQLI is to strongly emphasize on ‘how to’ implement
Since long time QbD have been used for various products
guidance according to ICH-Q8, Q8 (R), Q9 and Q10 [9].
development like Design of Experiments is being used since
Hence, QbD for pharmaceuticals is a systematic, scientific,
1920’s as factorial designs mainly in agricultural sciences,
risk-based, complete approach to develop the best optimized
later in fifties the technique was more accepted and became
pharmaceutical dosage form. Designing and development of
popular for industrial applications. FMEA (Failure Modes
formulations and manufacturing processes with help of QbD and Effects Analysis) was developed 1950s to study prob-
ensures predefined product quality objectives. QbD is also
lems that arise from malfunctions of military systems. An
useful in identification of characteristics (Fig. 2) that are
FMEA is often the first step of a system reliability study.
critical to quality. QbD analyze the effect of critical process
With further development in 1990 software were developed
parameters i.e. independent variables and indicates how
in the QbD methodology.
these important and specific process variables should be

'UXJ

ĐĐĞƉƚĂŶĐĞ
ƌŝƚĞƌŝĂ>ŝŬĞ WƌŽĚƵĐƚ
ůůhŶŝƚ /ŶͲƉƌŽĐĞƐƐ
ƐƐĂLJ͕ ^ƉĞĐŝĨŝĐĂƚŝŽŶ
KƉĞƌĂƚŝŽŶƐ ^ƉĞĐŝĨŝĐĂƚŝŽŶ YƵĂůŝƚLJ
ŝƐƐŽůƵƚŝŽŶ͕
ĨŽƌsĂƌŝŽƵƐ WƌŽĚƵĐƚ
^ŽůǀĞŶƚ
WƌŽĐĞƐƐ
ZĞƐŝĚƵĞ
dĞƐƚŝŶŐ

,IIDLO ,IIDLO

([FLSLHQWV 0DWHULDO 3URGXFW1RW


'LVFDUGHG $FFHSWHG

Fig. (2). Quality by testing.


4 Drug Delivery Letters, 2015, Vol. 5, No. 1 Singh and Sharma

Because of these techniques for getting better product (CPPs) and Critical Quality Attributes (CQAs) in
rather best product the techniques were more highlighted in such a way that enables the operational criteria to be
the pharmaceutical fields. USFDA report on Pharmaceutical contained within a Design Space.
cGMP for 21st century results in a strategic changes and
involvement of more statistical approach in any new QbD for Pharmaceutical Products - Important Steps in
development or for betterment of the old products also, its Implementation [17]
thereby laying the groundwork for QbD methodology (Fig. 3).
The same ground further fruits in development of new Identification of Target Product Profile Quality character-
guidance document from FDA, i.e. process analytical technology istic means prospective and dynamic summary of the quality
“PAT — A Framework for Innovative Pharmaceutical characteristics of a drug product that ensures the desired
Development, Manufacturing, and Quality Assurance”. quality, safety & efficacy.
Although the focus was more geared towards PAT, this
guidance document discussed many principles of QbD. In 1) First and foremost is to identify CQAs i.e. any
2005, the concept of design space was introduced in physical, chemical, biological, or microbiological
Common Technical Document (CTD) of ICH-Q8. This property or characteristic.
Design Space was basically a method which concentrates on 2) Defining and generating Process Design Space i.e.
differentiation between product attributes and process performing risk assessment linking material attrib-
parameters especially critical variables on which the focus utes and process parameters to CQAs.
should be given. The approach used to decide the critical
variables is risk assessment. An ICH Q9 guideline includes 3) Establishing Design Space Linkage between input
the tools that can be used to validate risk assessments and to independent variable, process parameter and CQAs.
check risks that have been identified. ICH-Q10 is the next 4) Defining the Controlling methodology as planned
approach, which address, development and manufacture of set of controls, derived from current product and
Pharmaceutical drug substances systems. These guidelines process understanding that assures process perform-
apply to pharmaceutical drug substances and drug products, ance and product quality.
as well as biotechnology and biological products.
5) Process Validation involves understanding of the
Lastly ICH-Q8 (R) gives details of the principles of QbD manufacturing process.
and clarifies the key concepts described in ICH-Q8. The aim
of the annex is to show how concepts and tools could be 6) Process monitoring is the process that is performing
used in practice by the applicant for all dosage forms. Both within the defined acceptable variability that served
the guidelines, ICH-Q10 and Q8 (R) are still subjected to as the basis for the filed process design space.
revision [15]. Overall these steps may be summarized as [18]:
QBD involves the following essential key aspects/elements
during pharmaceutical development:- A. Ascertaining Drug Product Objectives
1) Explain target product quality profile. A quality target product profile QTTP is embarked upon
encompassing the fundamental information of the product to
2) Designing and development of product and
be prepared or aspired as “goal-setting” exercise through
manufacturing processes.
brain storming among the team members cutting across in-
3) Recognition of critical quality attributes, process dustrial disciplines.
parameters, and variability sources.
4) Lastly controlling the manufacturing processes to B. Prioritizing Input Variables for Optimization
get consistent quality over time and so on. The critical material attributes CMA and critical parame-
ters CPP, which directly influence the CQA, represent the
Technical Elements of the QbD Process product quality, are analyzed through the Ishikawa Fish dia-
QbD process consists following technical elements as gram and prioritization studies.
shown in figure 4: [16].
C. Design-guided Experimentation and Analysis
1) Critical Quality Attributes (CQAs) are very much
connected to clinical relevance because of their im- Response surface optimization carried out employing
pact on efficacy, safety, or reproducible therapeutic experimental design is considered as a pivotal part of QbD
effect. exercise. The experimental design helps in mapping the re-
sponses on the basis of the studied objectives, CQA being
2) Clinically relevant CQAs which is linked to critical explored, at high, medium or low levels of CMA.
process parameters (CPPs), either directly or indi-
rectly. D. Modelization and Validization of QbD Methodology
3) One or more CPPs, which control the clinically rele- RSM is employed to relate a response variable or CQA to
vant CQA enabled by real time monitoring or by PAT. the levels of input variables or CMAs/CPPs using the inter-
4) Acceptance Criteria which is defined by dimensional play of polynomial equations, the desired constraints/criteria
relationships between critical process parameters for optimum search and the design constraint.
Quality by Design (QbD) Approach in Pharmaceuticals: Status, Challenges and Next Steps Drug Delivery Letters, 2015, Vol. 5, No. 1 5

Fig. (3). Typical layout of QbD.

Fig. (4). Elements of QbD.

E. QbD Validation, Scale-up and Production Target Product Quality Profile


Validation of the QbD methodology is a crucial step that The target product quality profile (TPQP) is an essential
forecasts about the prognostic ability of the polynomial element of QbD approach. It provides an understanding of
models studied. Qbd performance is corroborated after scal- product quality, safety, and efficacy. TPQP is useful and
ing up the results obtained from validation to ensure the re- used in designing and optimization of a formulation and
producibility and robustness. manufacturing process. ISPE and PQLI also call this the
Various measures taken for assuring all of the above Pharmaceutical Target Product Profile [21, 22].
summarized steps A-E are-
Critical Quality Attributes
Target Product Profile As per ICH, CQA is define as quality attribute that com-
FDA gives guidelines for defining Target Product Profile prises of physical, chemical, biological or microbiological
(TPP) [19]. As per these guidelines “The TPP is totally cor- characteristic and it is desired that these attributes should be
related to drug development program that provides knowl- to be controlled (directly or indirectly) to give assurance that
edge of drug during development. Generally, the TPP is use- product obtained will attain desired safety, efficacy, stability
ful to develop a link for drug labeling, drug development and performance as well [23].
activities as well as to some concepts that are useful in the ICH-Q8 revision ICH-Q8 :( R1) states that all the above
drug labeling”. According to ICH-Q8 (pharmaceutical de- mentioned quality attributes should be within appropriate
velopment), pharmaceutical development should include limit. CQAs give the assurance of product of desired quality
“Recognition of critical quality attributes of the drug product [24]. Elements of the TPQP can be described by CQA like
with consideration of its intended use as well as route of ad- dissolution. In determination of product performance CQA is
ministration”, hence it becomes essential to consider the in- used to observe the mechanistic variables such as particle
tended usage and route of administration [20]. size and hardness. Hence both the aspects i.e. product per-
6 Drug Delivery Letters, 2015, Vol. 5, No. 1 Singh and Sharma

formance and product performance determinants, could be ucts. Each and every stage of product lifecycle should be
explained by using TPQP [25, 26]. validated by using elements of Q10 in an appropriate and
proportionate manner. Principles of ICH-Q10 also used to
Comparison between End Product Testing and QbD recognize the differences, and the different objectives of
each stage. These guideline applies in the process that help in
Testing of end product is used to confirm the quality of design, development and preparation of drug substances such
the formulated product although it does not help to maintain as API and drug products which includes biotechnology and
the consistency of the formulated product as well as process biological products, throughout the lifecycle of product [32].
control while in case of QbD product quality is ascertained
by observing the data from a number of small batches that
are supposed to be acceptable and fall under desired accept-
able limit [27, 28].

ICH Guidelines
• Pharmaceutical Development (Q8)
ICH-Q8 is intended to provide guidelines of Pharmaceu-
tical Development of drug products as explained in the scope
of Module 3 of the CTD. While clinical trial stage of the
product the guidelines of ICH-Q8 are not applicable for the
data at the same time the principles in these guidelines are
important to consider clinical trials. ICH-Q8 guidelines are
also suitable for various other products but the application is
only possible after taking due approval from the appropriate
regulatory authorities. The annex provides further clarifica-
tion of key concepts outlined in the core Guideline. In addi- Fig. (5). Quality attributes governing quality of desired product.
tion, ICH-Q8 (R1) states that the annex explains the princi-
ples of quality by design (QbD) and this annex is not ment to
generate new standards but explains the concepts and tools
and their application by applicant in design space, outlined in
the parent Q8 document [29].

• Quality Risk Management (Q9)


ICH-Q9 Guideline is mainly concerned with the princi-
ples and examples of quality risk management (QRM). In
other word ICH-Q9 may be regarded as the document having
answers to all questions related with quality of product i.e.
parameters including designing, development, processing,
distribution, inspection/assessment and submission/review
processes throughout the lifecycle of drugs, drug products as
well as biological and biotechnological products, also in-
cludes raw materials, vehicles/solvents, additives and mate-
rial used for packing as well as labeling [30]. Risk assess- Fig. (6). Conventional versus QbD manufacturing process.
ment is one of the essential step require for ensuring the ef-
fective risk management. It reviews the materials, processes,
Noncritical Parameters
equipment used, storage conditions and intended use of the
product. The parameters used in the design space generation, are
In the risk assessment some of the questions like are ad- all critical but these not applied equally on the edges of de-
sign space and here comes some of the noncritical parame-
dressed:
ters which may not be captured in the design space, also pre-
Most vulnerable step which might go wrong? What is the sent ambiguity. Although noncritical process parameters are
probable nature and chances of occurrence, of risk? What not responsible for the failure in target product quality pro-
may be the results because of risk? etc. All of these decide file (TPQP) in the potential operating space and cause no
the step which should be addressed during QRM activities interactions with other parameters in the proven acceptable
for better control on the complete development process of range. Noncritical parameters can be monitored and con-
the product [31]. trolled easily [33, 34].

Pharmaceutical Quality System (Q10) Difference between Conventional Approach and QbD
ICH-Q10 Guideline describes quality of drug substances Approach
and its products (Fig. 5), which includes biotechnology and In case of Conventional approach, assurance of quality is
biological products, throughout the lifecycle of drug prod- subjected to its testing and inspection. It includes thorough
Quality by Design (QbD) Approach in Pharmaceuticals: Status, Challenges and Next Steps Drug Delivery Letters, 2015, Vol. 5, No. 1 7

Table 1. Comparison of the current state to the future desired QbD state.

Criteria Current state QbD state required

Pharmaceutical development Empirical; typical single variable experiments Systematic; multivariate experiments

Manufacturing process Rigid: avoids changes Flexible: changes can be made in design

By in-process testing
Process control Either in-process quality checks (IPQC) or off-line finished By using PAT for feedback and feed forward in real time
product quality checks

Based on performance of product in quality control


Product specification On behalf of previous experiences and batch data
checks

Control strategy Mainly by IPQC and end product testing Risk based controls shifted up-stream; real time release

Response after getting problem in IPQC or End product Design space itself have the provision of continual im-
Lifecycle management
testing, i.e. changes after approval are needed. provement of product during its development

data submission which includes coherent knowledge. All the sion of the product or process as well as to acquire an excel-
specifications are provided on behalf of previous experiences lent and economic product.
i.e. batch history. Conventional approach is rigid and always
avoids changes. It focuses on reproducibility which often CONFLICT OF INTEREST
avoids or ignores variation.
The author(s) confirm that this article content has no con-
While in QbD approach, attempt is made to develop qual- flict of interest.
ity in the product and process by help of design (any) which
depend upon scientific knowledge of product and process. ACKNOWLEDGEMENTS
Here, specifications are based on performance of the product
required. This approach is a flexible approach and changes The authors are thankful to Shri Dev Murti Ji, honorable
can be made in design for product quality improvement. It chairman, SRMS trust, Bareilly (U.P.) for inspiring us and
focuses on lustiness to understand as well as to control the providing all necessary facilities.
variables (Fig. 6) [35].
REFERENCES
Nasr summarized the differences between ‘‘current
[1] Food and Drug Administration. Final Report on Pharmaceutical
state’’ and ‘‘desired state’’ as shown in Table 1 [36]. cGMPs for the 21st Century–A Risk Based Approach. Available
from: http://www.fda.gov/ cder/ gmp/ gmp 2004/GMPfinalreport,
Software Used for Qbd 2004.html [Accessed May 10, 2014].
[2] U.S. Department of Health and Human Services Food and Drug
Software available for QbD includes Design Expert®, Administration Pharmaceutical cGMPS for the 21st Century – A
MODDE®, Unscrambler®, JMP®, Statstica®, minitab® etc, Risk-Based Approach: Second Progress Report and Implementa-
are at the software usually provide interface guide at every tion Plan, 2004.
[3] Pharmaceutical Quality for the 21st Century: A Risk-Based Ap-
step during the entire product development cycle. Software proach. Available from: http://www.fda.gov/oc/cgmp/report0507.
provides support for chemo-metric analysis through multi- html [Accessed May 10, 2014].
variate technique like MNLRA, PCA, PLS etc. [18]. [4] McCurdy, V. Quality by Design: Process Understanding, For
Scale-Up and Manufacture of Active Ingredients, 1st ed.; Wiley-
CONCLUSION VCH Verlag GmbH & Co. KGaA, 2011; pp. 1-16.
[5] Early, F.J.; Quality by Design, Part 1 Preventing quality failures at
Now a days researcher utilizes QbD as an important tool their source. Available from: http://www.qualitydigest.com/inside/
quality-insider-article/quality-design-part-1.html [Accessed May
for getting the quality, QbD has eliminated the burden of 10, 2014]
exhaustive and time consuming conventional approach with- [6] Juran, J.M. Juran on Quality by Design: The New Steps for Plan-
out compromising with quality of the product. Hence QbD is ning Quality into Goods and Services, appendix to chapter 3, The
an integral part of modern research in pharmaceutical indus- non-Pareto principle; mea culpa. New York: The Free Press, 1992;
try. All the concepts of ICH guidelines whether they are re- pp 68-71.
[7] US Food and Drug Administration. Guidance for industry: Q8
lated to product development, quality risk management or to Pharmaceutical Development, US Department of Health and Hu-
pharmaceutical quality system, are essential for processing a man Service, FDA, Rockville, MD, May 2006.
pharmaceutical process in QbD. QbD focus over all aspects [8] Sandipan, R. Quality by Design: A Holistic Concept of Building
desired in an quality product like ascertaining drug product Quality In Pharmaceuticals. Int. J. Pharm. Biomed. Res., 2012,
3(2), 100-108.
quality profile, priorizing input variables for optimization, [9] Drennen III, J.K. Quality by design-what does it really mean? J.
modelization & validation of QbD methodology and in the Pharma. Innov., 2008, 2, 65-66.
last QbD validation, scale up and production as well as soft- [10] Moheb, M. Nas. Implementation of Quality by Design (QbD) –
ware used for QbD. Hence we can say that QbD is a useful Current Perspectives on Opportunities and Challenges Topic Intro-
tool in the pharmaceutical research to facilitate comprehen- duction and ICH Update, Office of New Drug Quality Assessment
8 Drug Delivery Letters, 2015, Vol. 5, No. 1 Singh and Sharma

OPS/CDER/FDA Advisory Committee for Pharmaceutical Science http://www.ich.org/LOB/media/MEDIA4349.pdf [Accessed Febru-


and Clinical Pharmacology. ary 20, 2014].
[11] Yu, L.X. Pharmaceutical quality by design: product and process [25] Nosal, R. PQLI-criticality. ISPE PQLI Berlin Conference. Sept.
development, understanding and control. Pharm Res., 2007, 25(4), 2007.
781-791. [26] Tong, C.; D-Souza, S.S.; Parker, J.E.; Mirza, T. Commentary on
[12] Singh, L.; Nanda, A.; Sharma, S.; Sharma, V. Performance Optimi- AAPS workshop dissolution testing for the twenty-firstcentury:
zation of Buoyant Beads of Anti-Diabetic Drug Using Quality by Linking critical quality attributes and critical process parameters to
Design (QbD). Latin Amer. J. Pharm., 2014, 33, 14-23. clinically relevant dissolution. Pharm. Res., 2007, 24, 1603-1607.
[13] Singh, L.; Nanda, A; Sharma, S.; Sharma, V. Design Optimization [27] Woodcock, J. The concept of pharmaceutical quality. Amer.
and Evaluation of Gastric Floating Matrix Tablet of Glipizide. Pharm. l Rev., 2004, 1-3.
Trop. J. Pharm. Res., 2013, 12(6), 1596-5996. [28] Food and Drug Administration, Office of Generic Drugs White
[14] International conference on harmonization. Harmonized Tripartite Paper on Question-based Review. Available from:
Guideline on Pharmaceutical Quality Systems Q10, step 4 version, http://www.fda.gov/cder/ OGD/QbR.htm [Accessed February 20,
dated June 4, 2008. 2014].
[15] Gracia, T.; Cook, G.; Nosal, R. PQLI key topics- criticality, design [29] Q8 (R1): Pharmaceutical Development, Revision 1, ICH Harmo-
space and control strategy. J. Pharm. Innov., 2008, 3, 60-68. nized Tripartite Guidelines, International Conference on Harmoni-
[16] PQRI-FDA Workshop on Setting Drug Specifications for the 21st zation of Technical Requirements for Registration of Pharmaceuti-
Century, 1342 Bethesda, MD, March 16-18, 2005. cals for Human Use, 2007.
[17] Lawrence, X. AAPS annual meeting and exposition; Los Angeles [30] Q9: Quality Risk Management. ICH Harmonized Tripartite Guide-
convention centre, Los Angeles, CA, USA, 2009. lines. International Conference on Harmonization of Technical Re-
[18] Singh, B.; Beg, S. Quality by design in product development life quirements for Registration of Pharmaceuticals for Human Use,
cycle. Chron. Pharmabiz., 2013, 22, 72-79. 2006.
[19] FDA CDER. Draft guidance for industry and review staff. Target [31] Frank, T.; Brooks, S.; Creekmore, R.; Hasselbalch, B.; Murray, K.;
product profile—a strategic development process tool. March, Obeng, K.; Reich, S.; Sanchez, E. Quality risk management princi-
2007. Available From: http://.www.fda.gov/downloads/drugs/ ples and industry case studies, sponsored by the Product Quality
guidancecompliancereguatoryinformation/guidances/ucm.080593 Research Institute Manufacturing Technology, Committee, De-
[Accessed February 20, 2014]. cember 2008. (PQRI-MTC) (http://www.pqri.org).
[20] FDA CDER. Guidance for industry. Q8 Pharmaceutical development. [32] Q10: Pharmaceutical Quality System, ICH Tripartite Guidelines.
May2006.http://www.fda.gov/downloads/RegulatoryInformation/ International Conference on Harmonization of Technical Require-
Guidances/ucm128029.pdf [Accessed February 20, 2014]. ments for Registration of Pharmaceuticals for Human Use, 2007.
[21] Yu, L.X.; Raw, A.; Lionberger, R. U.S. FDA Question-based re- [33] Lionberger, R.A.; Lee, S.L.; Lee, M.; Raw, A.; Yu, L.X. Quality by
view for generic drugs: a new pharmaceutical quality assessment Design: Concepts for ANDAs. AAPS J., 2008, 10(2), 268-276.
system. J. Generic Med., 2007, 4, 239-248. [34] Laura, B. The role of noncritical process parameters in quality by
[22] ISPE PQLI. Draft PQLI summary update report. Available from: design. BioPharm. International., 2010, 23(10), 1-2.
http://www.ispe. org/cs/pqli product quality lifecycle implementa- [35] Bhatt, D.; Rane, S. QbD Approach to analytical Method Develop-
tion /draft pqli summary update report [Accessed February 20, ment and its Validation. Inter. J. Pharm. Sci., 2011, 3(1), 179-187.
2014]. [36] Nasr, M.M. Quality by design (QbD) –a modern system approach
[23] Berridge, J. ICH Q8 & Q9 (+Q10) defined and undefined: gaps and to pharmaceutical development and manufacturing – FDA perspec-
opportunities. ISPE PQLI Washington Conference. June 2007. tive. FDA Quality Initiatives Workshop, February 28, 2007.
[24] International conference on harmonization. Draft consensus guide-
line: pharmaceutical development annex to Q8. Available from:

Received: May 12, 2014 Revised: November 04, 2014 Accepted: November 12, 2014

View publication stats

You might also like