QBD Application For The Pharmaceutical Development Process

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 34

Journal of Pharmaceutical Investigation (2022) 52:649–682

https://doi.org/10.1007/s40005-022-00575-x

REVIEW

Quality by Design (QbD) application for the pharmaceutical


development process
Sang‑Ho Lee1 · Jin‑Ki Kim2 · Jun‑Pil Jee3 · Dong‑Jin Jang4 · Young‑Joon Park5 · Joo‑Eun Kim1,6

Received: 27 August 2021 / Accepted: 26 April 2022 / Published online: 29 August 2022
© The Author(s) under exclusive licence to The Korean Society of Pharmaceutical Sciences and Technology 2022

Abstract
Background Quality by Design (QbD) is a pharmaceutical quality management system that predicts, controls, and system-
atically responds to risks that may occur in the research stage and manufacturing process. There are many articles on how
to improve quality through application of the QbD system, but the methodology surrounding how to specifically apply this
process is limited. The purpose of this review was to provide accurate information on drug development process and overcome
the difficulties of developing QbD-applied drug products through an appropriate application method.
Area covered QbD-based pharmaceuticals are continuously being developed worldwide, helping to identify potential risks,
and managing all aspects, from drug design to patient consumption, further enabling the patient to be administered a high-
quality drugs based on their design. In this study, a basic knowledge of QbD, its application and effectiveness during drug
development will be discussed.
Expert opinion Our results suggest that a QbD for drug development can reduce risk to patients, improve drug quality, and
continuously produce superior quality drug products.

Keywords Quality by Design · Pharmaceutical development process · QTPP · CQAs · CMAs · CPPs

Introduction health, safe and effective medicines are required (Patel et al.
2013). In 1958, the American Pharmaceutical Industry
Quality control is essential to ensure the safety and efficacy of Association formed the Quality Assurance Committee in
pharmaceuticals (Jin et al. 2014). Due to the safety and effec- response to the increasing frequency of compensation cases
tiveness of drug products and their direct effects on patient for damages caused by defective drugs in the United States of
America. After the thalidomide drug accident in 1961, good
* Young‑Joon Park manufacturing practice (GMP) was enacted in 1962, and the
parkyj64@gmail.com
Food and Drug Administration (FDA) GMP was adopted
* Joo‑Eun Kim in 1963. Thus, the historic efforts by several countries to
77jooeun@naver.com
ensure the quality of pharmaceuticals and secure interna-
1
Department of Pharmaceutical Engineering, Catholic tional competitiveness extends back multiple decades, and
University of Daegu, Hayang‑Ro 13‑13, Gyeongsan, enormous resources have been invested. For example, there is
Gyeongbuk 38430, Republic of Korea the pharmaceutical inspection co-operation scheme (PIC/S)
2
College of Pharmacy & Institute of Pharmaceutical Science (Kim and Kwon 2013; Uhlenbrock et al. 2017), which is an
and Technology, Hanyang University, 55 Hanyangdaehak‑ro, international consultative body that is tasked with creating a
Sangnok‑gu, Ansan 15588, Republic of Korea
global consensus of pharmaceutical manufacturing, quality
3
College of Pharmacy, Chosun University, Pilmun‑daero 309, control standards, and GMP inspections (Kothari and Patel
Dong‑gu, Gwangju 61452, Republic of Korea
2015; Ban et al. 2019). The primary responsibility of PIC/S
4
Department of Bio‑Health Technology, College is to conduct various activities to manufacture and manage
of Biomedical Science, Kangwon National University,
Chunchoen 24341, Republic of Korea high-quality medicines, such as mutual exchange of basic
5
GMP inspection information, coordination of GMP regula-
College of Pharmacy, Ajou University, 206 World cup‑ro,
Yeongtong‑gu, Suwon 16499, Republic of Korea
tions, publication of guidelines, training of GMP inspec-
6
tors, and regular meetings; however, despite these efforts,
Department of Biopharmaceutical Chemistry, Kookmin
University, Seoul 02707, Korea
various quality problems persist. Pharmaceutical drugs are

13
Vol.:(0123456789)
650 Journal of Pharmaceutical Investigation (2022) 52:649–682

manufactured and quality controlled using the Quality by quality assurance standard in the global pharmaceutical mar-
Testing (QbT) technique, which focuses on risk management. ket. In addition, many international regulatory agencies and
As quality control through QbT is performed by selecting pharmaceutical companies have already completed the prepa-
manufactured drug products at random, there is an increased rations and conditions for introduction, having entered the full-
risk of supplying drugs with poor quality (Kader 2016; scale implementation phase. However, many pharmaceutical
Fukuda et al. 2018; Suciu et al. 2018). The current qual- companies are having difficulties in setting the developmen-
ity process capability index of the pharmaceutical industry tal directions due to a shortage of professional experts. As
is only 2–3σ. The yield rate for 2σ and 3σ is 95.45% and it incorporates quality management throughout the entire life
99.73%, respectively. The pharmaceutical industry spends cycle, from the initial stage of drug development, it is diffi-
5–10% sometimes up to 20% annually on handling defective cult to discard the existing paradigm and adopt QbD concepts
products. This was a huge amount equal to 20% of the annual over a short period. Beyond simple pharmaceutical product
sales. If there were no problems with the product process, development, risk factors should be evaluated in advance, and
the company's net profit would increase immediately. If the a control strategy should be pre-established using statistical
'lifecycle management' is pursued by QbD and the quality is techniques and advanced, real-time manufacturing analyses
improved step by step, a yield of 6σ (99.9999998%) can be to understand the assessed risks more. Further, one compo-
derived. This is only two to three defective products out of nent requires a vast amount of information to be applied to the
1 million. This is very effective for developing high-quality production of pharmaceutical products to guarantee quality.
pharmaceuticals. In comparison, the defect rate of 2–3σ in In the literature, there have been several studies to improve
the pharmaceutical industry is very high. quality through the QbD system, but the methodology for drug
To address these issues, the US FDA sought to apply the development is limited. Further, a study on the implementation
Quality by Design (QbD) technique as a quality management method and procedure for QbD application in pharmaceuti-
system to pre-emptively predict and manage possible risks cal development has been published (Chen 2006; Guideline
in the research stage and manufacturing process of drug 2008; Karanakov et al. 2012; Calnan et al. 2013; Aksu and
products, and systematically cope with them (Rathore and Mesut 2015; Peltonen 2018). However, research might be
Winkle 2009; Patil and Pethe 2013; Lawrence et al. 2014; hard to understand for those new to this field, and few the-
Waghule et al. 2019). QbD was first defined by Juran and was ses comprehensively deal with the easy application of QbD
designed to secure the quality of drug products from plan- in the development process of drugs. In a survey of actual
ning or design, not evaluation or control (Pharm and Pharm formulation, most researchers of the pharmaceutical industry
2013; Jain 2014; Kadam et al. 2017; Jagan et al. 2021). The answered that the introduction of QbD was necessary. How-
conceptual QbD approach in the pharmaceutical industry, ever, they stated, although they recognised the need for QbD,
often considered twenty-first century GMP, was introduced it was difficult to apply QbD in drug development, further it
by the US FDA in 2002 through a final report of ‘Pharma- was difficult to understand. The need for QbD is emphasised,
ceutical current good manufacturing practice (cGMP) for the but many pharmaceutical companies face difficulties applying
twenty-first century—a risk-based approach’. Furthermore, QbD because there are few papers with detailed descriptions
the FDA Guidance for Industry on Process Validation, first of specific implementation methods and procedures.
published in 1987, was revised in January 2011 to incorpo- Therefore, the present research examines the most appropri-
rate QbD principles (Bhutani et al. 2004; Beg et al. 2019); ate methods for applying QbD to the drug development pro-
in particular, to help identify the factors affecting critical cess and examine its benefits to the pharmaceutical industry
material attributes (CMAs), and critical process parameters and research scientists.
(CPPs) that can affect critical quality attributes (CQAs) of
the pharmaceutical product. Overall, QbD finds solutions to
problems during phenomenon of departure, manages risks, Comparison of conventional to QbD
and enables pharmaceutical production of uniform quality. development processes
The GMP manufacturing process and quality management
are operated as individual systems; but the most critical differ- In the development of conventional, orally administered
ence of QbD is that real-time management is possible because drugs, data is obtained on the active pharmaceutical
it is integrated into a single system (Chen 2006; Guideline ingredients (API) through a literature search, as well as
2008; Karanakov et al. 2012; Calnan et al. 2013; Aksu and performing API solubility tests, physico-chemical assays,
Mesut 2015; Peltonen 2018). The application of QbD is imper- and preformulation studies. Next, through a compat-
ative in drug development (Schweitzer et al. 2010; Grangeia ibility study among API, excipients that can and cannot
et al. 2020; Kajiwara et al. 2020), and as many countries be used are selected in advance before the formulation
demand advancement through QbD-based pharmaceutical research. Subsequently, various compositions can be made
development and management, it is becoming an essential through formulation research, and it is possible to select

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 651

the optimal manufacturing method among wet granulation, and the optimal quality is achieved. Similarly, the optimal
dry granulation, and direct compression through manufac- quality process and materials should be applied during
turing process research. drug development. The manner of the drug development
Approximately, three to five formulations are deter- process applying such QbD will be explained in the next
mined, besides evaluating the dissolution, disintegration step.
time, hardness, and friability of determined tablets, and
the optimal prescription quantity is confirmed within a
short time through a stability study of downscale methods. Quality by Design (QbD)‑based drug
When the lab scale formulations are complete, the prod- development
uct quality is evaluated through a pilot-scale composition
study and an in-process control; whereas the stability of Quality target product profiles (QTPPs)
the drug product is confirmed through a long- and accel-
erated-term stability test and accelerated condition for The initial step of the QbD is to establish the QTPPs, which
six months. Simultaneously, a non-clinical beagle phar- requires a clear understanding of target product profile (TPP)
macokinetics study of the drug product is conducted to of drugs. The QTPP summarises the predictive quality char-
confirm the in vivo pharmacokinetics such as the absorp- acteristics of a drug product to be achieved to ensure the
tion, distribution, metabolism, and excretion (ADME), and targeted quality standards of safety and efficacy are met
when appropriate, an investigational new drug applica- (Rathore 2009; Marto et al. 2015; Finkler and Krummen
tion (IND) for clinical trial is submitted to the appropriate 2016). By establishing QTPPs, goals and directions are pre-
regulatory agency. set before product development, as is the basis for pharma-
In QbT, the conventional pharmaceutical manufactur- ceutical formulation design (Purohit and Shah 2013; Gan-
ing quality control method, after manufacturing the drug dhi and Roy 2016; Kan et al. 2014; Thoorens et al. 2014).
through a standardised process, the batch results (interme- Broadly, QTPPs can be thought of as the contents of the
diate process materials, raw materials and finished drug paper insert included when purchasing a prescription drug;
product) are analysed and quality control analyses are per- however, they must be interpretable from the perspective
formed based on the results (Fig. 1). QbT relies on experi- of the pharmacist, patient, and the doctor to comprehen-
ence throughout the development, and conducts R&D on sively convey risks and justification. The QTPP compo-
a single variable (Bhusnure et al. 2015). In addition, it is nents of synthetic drugs primarily include indications, dos-
unfeasible to evaluate the quality of all pharmaceutical age, route of administration, pharmacokinetic profile, shelf
products, so a small sample is randomly selected from the life, packaging, etc. (Zurdo et al. 2015; Gupta and Jhawat
population following manufacturing completion, and the 2017); whereas those for biopharmaceuticals include trans-
level of defective products is relatively high (Bhoop 2014; parency, particulates, pH, activity, sterility, stability, colour,
Arora et al. 2016). odour, moisture content, dissolution time, lyophilised cake
However, to develop a pharmaceutical drug product to properties, etc. (Cunha et al. 2020a; Kanwal et al. 2021).
which QbD is applied, one must first define and verify QTPP can be defined more clearly by pre-judging the risk
the development quality target product profile (QTPP) for each pharmaceutical in consideration of its character-
and set the CQAs based on prior knowledge and experi- istics and development setting. Further, understanding the
ence according to the stated purpose (Fig. 1). Then, an needs of patients to manage their disease is helpful in select-
extracting process of formulation variables and process ing a QTPP. For example, the risk level must be set to low
parameters is required, and by using design of experimen- when selecting a natural oral route of administration during
tal (DoE) techniques, the results obtained that satisfy the manufacturing, and the justification must also state that oral
CQA criteria are screened using statistical methods, and administration is the route of ease with the patient’s compli-
preliminary analyses of risk factors that may occur during ance. In addition, the target value of the pharmacokinetic
pharmaceutical manufacturing are conducted. CMAs and item is a further risk to be considered, as it can affect clinical
CPPs with high risk and impact on CQA are identified, results such as bioequivalence, safety, and efficacy of the
and for the data obtained that satisfy the CQA criteria in active pharmaceutical ingredient, and any justifications must
a robust and reproducible manner, the relationship and consider this (Table 1).
interactions between these quality variables, CMA, and
CPP are modelled; thus, it is necessary to verify and build Critical quality attributes (CQAs)
a design space. Finally, by controlling the formulation
and process variables within the design space, as well as The CQAs indicate that a physical, chemical, (micro) bio-
establishing and executing an optimised control strategy logical characteristic, or other properties are within reason-
to consistently satisfy the CQAs, their risks are minimised able distributions to ensure the desired quality. CQAs are

13
652 Journal of Pharmaceutical Investigation (2022) 52:649–682

Fig. 1  a Conventional Quality by Testing (QbT) for drug develop- with permission of Zhang and Mao, Asian Journal of Pharmaceutical
ment, and the b QbT, c Quality by Design (QbD), and d QbD-based Sciences, ScienceDirect, 2016.)
drug development approaches (Zhang and Mao 2017). Reproduced

13
Table 1  Example of quality target product profile (QTPP) definition
QTPP Target Risk Justification Related quality attributes References

Indication Ex) Reflux esophagitis Yes or No Inconsistent indications for patients tak- Appearance Raw et al. (2011), Finkler and Krummen
ing this drug as a therapeutic drug may (2016), Bhambure et al. (2011)
cause unnecessary drug misuse and
side effects to the patient. Therefore,
the indications for the drug should be
clearly explained
Dosage design Ex) Coated tablets of not more than 160 Yes or No Since the dosage form is one of the Appearance Namjoshi et al. (2020)
mg most important elements of the Drug
Ex) Transparent medical ampoule with Delivery System(DDS), it is directly
yellow liquid drug solution related to the drug efficacy. Therefore,
the concept and justification of the for-
mulation should be clearly described
Route of administration Ex) Oral, Injection, transdermal, sublin- Yes or No The route of administration is directly Test Items Gavan et al. (2017a), Syed and More
gual tablet related to patient compliance. Also, (2020)
Journal of Pharmaceutical Investigation (2022) 52:649–682

errors in the route of administration


may affect drug efficacy. For example,
in the case of a sublingual tablet, if it
is taken like a regular tablet, it will not
be able to show its proper medicinal
effect. Therefore, the route of adminis-
tration should be clearly described
Dosage strength Dosage Yes or No When administered in a certain amount Assay Djuris et al.( 2013), Hasnain et al. (2021),
or more, side effects may occur, and Mukharya et al. (2012a), Marto et al.
if administered in less than a certain (2016)
amount, the therapeutic effect may be
reduced. Therefore, it is necessary to
establish a dosage strength that can
maintain an appropriate treatment area
and prove its validity
Pharmacokinetics AUC/Cmax/Tmax Yes or No The pharmacokinetic profile can directly Assay Gavan et al. (2017b), Gavan and Porfire
affect safety and efficacy. Therefore, it (2017a), Charoo et al. (2017), Shah et al.
is required to establish a pharmacoki- (2017)
netic profile that can prove safety and
efficacy through various non-clinical
tests
Stability and Stable for a period under storage condi- Yes or No The quality attributes of the drug Assay Patel and Parmar (2013), Mesut et al.
shelf life tions must be suitable for the set period, Impurities (2015)
otherwise, the drug cannot be properly
effective. Because the shelf life of a
drug directly affects safety and effec-
tiveness, it must be clearly indicated
and the rationale for the setting must
be demonstrated

13
653
Table 1  (continued)
654

QTPP Target Risk Justification Related quality attributes References

13
Appearance Appearance of the target formulation Yes or No Changes in product appearance can Appearance Vivekanandan (2019), Dangre et al.
lead to errors in the patient group’s (2019), Hasnain and Ahmed (2021)
selection of products for treatment.
Therefore, the appearance must be
clearly described (engraving, color,
shape, etc.)
Moisture content Not more than ~ % Yes or No Moisture may have a cause such as Moisture content Hasnain and Ahmed (2021), Cunha et al.
lowering of dissolution and increase (2020b)
of impurities. Therefore, it is neces-
sary to clearly establish the allowable
moisture range and justify it for proper
management
Identification Same peak retention time Yes or No The API must be identified. Absence of Identification Chordiya et al. (2019), Mundada et al.
API should be managed as it cannot be (2017)
effective
Assay 95–105% Yes or No If the assay is high, side effects may Assay Nayak et al. (2019), Singh et al., Gavan
occur, and if the assay is low, sufficient et al. (2017b)
drug effects may not be expressed.
Therefore, it is necessary to establish
an appropriate assay range and clearly
explain its justification
Weight Meets pharmacopoeia criteria Yes or No Content uniformity may affect clinical Assay (rights are reserved by Sharmada and
variation/ trials, and it should be managed so that Kakodkar 2015), (Singh et al. 2010)
Content a consistent product can be manufac-
uniformity tured
Dissolution Dissolution pattern suitable for drug Yes or No Dissolution is important for bioavail- Dissolution Negi et al. (2016), Mishra and Raza
properties ability. Therefore, the target dissolution (2021), Vivekanandan (2019)
profile should be clearly described and
the in vitro in vivo correlation (IVIVC)
should be verified
Impurities Select considering the stability of the Yes or No As impurities directly affect the stabil- Impurities Abhinandana and Nadendla, Zhang and
API ity of the formulation, they must be Mao (2017), Desai (2018)
managed. Therefore, it is necessary to
set the permissible range of impurities
in advance and, if necessary, identify
the structure of impurities and manage
them
Residual Residual solvent Yes or No Residual solvents can deteriorate the Residual solvent Singh and Sharma (2015), Mandlik et al.
solvent Not more than ~ ppm properties of the formulation and (2020)
should be managed
Journal of Pharmaceutical Investigation (2022) 52:649–682
Journal of Pharmaceutical Investigation (2022) 52:649–682 655

associated with the drug substance, excipients, intermedi-

Purohit and Shah (2013), Mukharya et al.


ates (in-process materials) and the finished drug product.
Syed and More (2020), Patil and Pethe They include characteristics of quality: assay, uniformity
(2013), Kenjale et al. (2020) of dosage units, re-dispersibility, reconstitution time, and
aerodynamic property; safety: impurities related substances,

(2012b), DESIGN (2012)


residual solvents, osmolarity, isotonicity, microbiological
limits, sterility, and particulate matter; efficacy: diffusion,
dissolution, permeation; and multidisciplinary: patient
acceptance and compliance (Lawrence 2008). Because it is
Related quality attributes References

difficult for formulation researchers to understand the rela-


tively new subject of QbD, it is perhaps easier to understand
that it is the target result of the Certificate of Analysis (CoA)
report of the finished drug product; however, CQAs con-
tain an additional item asking whether it is a CQA, with the
corresponding justification written according to the target
standard. As CQAs are an important product characteris-
tic that can significantly affect the QTPP through the for-
Appearance
Impurities

mulation or process variables, a potential CQA should be


Assay

selected based on QTPP (Mandenius 2009; Rathore 2016).


For example, with a general oral tablet, the CQA items pri-
may react with drugs and promote their

to verify its justification and manage it

marily include descriptions such as appearance, identifica-


to select a packaging material that can
degradation. Therefore, it is necessary
surrounding environment. In addition,

guarantee the stability of the drug and


stability by protecting drugs from the
deterioration or degradation products
should be managed as they can cause

packaging materials in direct contact

tion test, assay, uniformity of dosage units, moisture content,


Yes or No In the case of microorganisms, they

Yes or No Packaging materials increase drug

impurities of related substances, residual solvent, and dis-


solution (Colobatiu et al. 2019). Furthermore, in the CQA
selection stage, one can more effectively attain the desired
quality of target drugs by judging and justifying the risks
of pharmaceuticals

for each quality attributes (QA) item, in consideration of


the characteristics of the drug substance and finished drug
Justification

product (Pramod et al. 2016). CQAs should be managed and


controlled throughout the product lifecycle management,
from delivery to the patient (Negi et al. 2015). For example,
with a generic oral tablet, the assay and content uniformity
test can be selected as a CQA because it indicates attributes
Risk

that can affect and compromise safety and efficacy; however,


since the identification test needs only to confirm the pres-
ence or absence of the active pharmaceutical ingredient, it
maintaining the physicochemical stabil-

does not have a significant effect on the formulation research


Packaging and container suitable for

and process parameters of the drug product (i.e., it is a gen-


Meets pharmacopoeial criteria

eral QA, not a CQA; Table 2a). The same can be said of a
general injection product, where a pH test can be selected
ity of pharmaceuticals

as a CQA; however, In addition, the aseptic and endotoxin


test can be selected as critical CQA because it is a quality
attribute that can affect and compromise safety. However,
the identification test would pertain to general QA, but not
CQA (Table 2b).
Target

Compatibility studies between API and excipients


Table 1  (continued)

The selection of excipients for the formulation step directly


Microbial limit

affects product quality; thus, the justification of the ration-


ale supporting the selection of excipients is critical. When
packaging
Primary

designing a drug product, compatibility tests should be


QTPP

performed on all excipients that may comprise part of the

13
Table 2  Example of critical quality attributes (CQAs) determination in: (a) tablets, and (b) injections
656

Quality Attributes Objective CQA Justification Can be replaced References


with other quality

13
attributes

(a)
Appearance It should be in a shape and color for Yes or No Color, shape, and appearance are not Yes or No Chaudhury et al. (2014), Yu et al. (2019),
patients’ directly linked to safety and effective- Ha et al. (2017)
convenience in taking, and as a tablet, no ness. However, patient compliance
defects to medication can be important for
should be observed patients to take medication. There-
fore, the appearance must be clearly
described and managed for patient com-
pliance to medication
Identification The main active pharmaceutical ingredi- Yes or No Identification directly affects safety and Yes or No Kolekar (2019), Su et al. (2019), Mishra
ent should efficacy. However, in the case of Iden- and Rohera (2017), McDermott et al.
be identified tification, it can be confirmed through (2015)
assay test, and since there are almost no
cases where API is not added, most of
it is written as No
Assay Ex) API 95.0–105.0% Yes or No Process variables can affect the content of Yes or No Soans et al. (2016), Freeman et al. (2015),
the drug product. Therefore, the content Wang et al. (2015)
should be evaluated through formula-
tion research and process development
Weight variation/ Meets pharmacopoeia criteria Yes or No Variation in content uniformity can affect Yes or No Su and Bommireddy (2019), Mohurle et al.
Content uniformity safety and effectiveness. That is, this (2019)
CQA should be evaluated through drug
product research and process develop-
ment. Otherwise, the efficacy of the
medicine may be affected
Moisture content No more than ~ % Yes or No Stability may be affected if the API is Yes or No Wang and Kan (2015), Chavan et al.
sensitive to moisture. Therefore, it is (2016)
necessary to clearly establish the allow-
able moisture range and justify it for
proper management
Impurities Select considering the stability of the Yes or No Impurities may affect safety. Therefore, it Yes or No Amasya et al. (2019), Dispas et al. (2018),
API should be evaluated through formula- Jovanovska (2018)
tion research and process development.
Also, if necessary, identify the structure
of impurities and manage them
Residual solvent Not more than ~ ppm Yes or No Residual solvents can affect safety and Yes or No Mesut and Ȍzsoy (2015), Wen and Park
stability depending on the physico- (2011), Shah et al. (2015)
chemical properties of the main compo-
nent, so it should be evaluated through
process development
Journal of Pharmaceutical Investigation (2022) 52:649–682
Table 2  (continued)
Quality Attributes Objective CQA Justification Can be replaced References
with other quality
attributes

Microbial limit Meets pharmacopoeia criteria Yes or No Failure to comply with the microbial Yes or No Amasya and Aksu (2019), Yamashita and
limit tests may affect patient safety. Sun (2020)
Therefore, a clear setting of the micro-
bial limit is required, and it must be
managed by justification its validity
Dissolution Dissolution pattern suitable for drug Yes or No The dissolution pattern can affect Yes or No Patil and Pethe (2013), Rawal et al. (2019),
properties bioavailability (efficacy). Therefore, Muntean et al. (2020), Kaushal et al.
the target dissolution profile should (2020)
be clearly described and the in vitro
in vivo correlation (IVIVC) should be
verified
(b)
Journal of Pharmaceutical Investigation (2022) 52:649–682

Appearance A colorless, transparent liquid for injec- Yes or No The appearance of the drug has a close relationship Yes or No Deng et al. (2019), Sonam and Rathore,
tion in a vial with the stability of the drug product. Stability is DESIGN (2012)
an important key quality characteristic in terms
of quality control, as it greatly affects safety and
effectiveness
Identification (thin layer chromatography) Yes or No Identification directly affects safety and efficacy. Yes or No Kenett and Kenett (2008), Stegemann et al.
R1 value ratio between standard solution However, in the case of Identification, it can be (2014), Silva and Silva (2021)
and test solution: 0.9–1.1 confirmed through assay test, and since there are
(Liquid Chromatography) almost no cases where API is not added, most of it Koeberle and Schiemenz (2017), Khurana
The standard solution and the sample is written as No et al. (2020)
solution should show the same reten-
tion time
Assay API 90.0–110.0% Yes or No The assay of the API directly affects the safety and Yes or No Amasya and Aksu (2019), Saitoh (2018),
effectiveness of the product, so it is one of the core Anderson et al. (2017)
quality characteristics that must be managed
pH 6.5–7.0 Yes or No It is one of the quality attributes that must be satisfied Yes or No Purohit and Shah (2013), Mandlik and
as an injection. In the case of pH, it may cause Dandgavhal (2020), Simões et al. (2018)
Osmotic pressure 280–340 mOsm Yes or No pain during injection. In addition, since it affects Soans and Chandramouli (2016)
the solubility of the drug, it should be managed by
setting an appropriate range and justification its
validity
Oxygen in vial Select later for the stability of the active Yes or No Oxygen in the vial can accelerate decomposition by Yes or No Alt et al. (2016), Saitoh (2018)
ingredient contact with the API, so it must be managed within
a certain limit. Therefore, if it is determined that
there will be changes according to the quantity
of product or manufacturing process variables, it
should be set as an output variable and managed

13
657
658 Journal of Pharmaceutical Investigation (2022) 52:649–682

final drug product (Fahmy et al. 2012) (Fig. 2). Excipients


with known safety risks, which are determined by compat-

Rathore and Mhatre (2011), Shah et al.

Thombre et al. (2021), Sri et al. (2021)


ibility tests, should be excluded from consideration during

Abhinandana and Nadendla, Chavda


the formulation step (Jacques et al. 1999). The exclusion
helps to ensure the stability of the drug product in advance

Santosh et al., Saitoh (2018)


Hasnain and Ahmed (2021) (Fahmy and Kona 2012). The Placket-Burman DoE is used

Dutta et al., Mistree et al.


Djuris and Ibric (2013)
to assess compatibility, as it can identify the most criti-
Sylvester et al. (2018)

cal factors early in the experimental process (Vanaja and


Shobha Rani 2007); therefore, this represents the optimal
design to confirm compatibility between API and excipients
References

(2009)

(2016)
(compatibility test examples are listed in Table 3). Mixed
samples of API and excipients are tested in various experi-
mental conditions, across different times and storage meth-
Yes or No

ods. The stability of the API is thus confirmed, dependent


with other quality

upon the presence or absence of specific excipients. When


Can be replaced

the compatibility test fails, the impurities increases rapidly


product. Therefore, if it is determined that there will

with time, and the endo- or exothermic peaks of differential


manufacturing process variables, it should be set as
attributes

be changes according to the quantity of product or


Yes or No directly affects the safety and effectiveness of the
Yes or No As it is an essential quality attribute of injection, it

scanning calorimetry (DSC) shift due to the degradation or


denaturation. Conversely, if the level of impurities is stable,
and there is no DSC peak shift observed, the compatibility
between the API and the excipient is considered good. The
Plackett–Burman design can confirm the interaction between
an output variable and managed

the API and a single, or multiple excipients, providing a


statistical basis for optimal selection (Verma et al. 2017).

Risk assessment

Risk assessment (RA) is described in the international coun-


Justification

cil for harmonisation (ICH) Guideline Q9. By performing


RA, the CMAs and CPPs that affect CQAs can be identified.
CMAs refer to the independent formulation variables, such
as physicochemical properties of active (drug substance)
Yes or No
Yes or No

Yes or No

Yes or No

Yes or No

and inactive ingredients (excipients) affecting the CQAs of


CQA

semi-finished and/or finished drug products. CPPs refer to


the independent process parameters most likely to affect the
There should be no discoloration inside

CQAs of an intermediate or finished drug product, as they


too should be monitored or controlled to ensure the desired
Foreign Insoluble Matter No detect foreign insoluble matter

quality. In addition, it also helps to set the research direction


in the early stages of development, and properly establish a
Not more than 0.17 EU/mg

design space (Kelley et al. 2016; Raina et al. 2017; Braem


No mycosis (negative)
6000 or less (per vial)

Not less than 2.0 m L


600 or less (per vial)

and Turner 2019). The RA tools that are available are listed
Not more than 15%

in Table 4 (ICH guideline Q9), where the potential variables


Insoluble visible particle 10 μg or more:

25 μg or more:

that can affect quality of the drug products are identified


the sample
Objective

(Pallagi et al. 2018; Borchert et al. 2020; Martinez‐Marquez


et al. 2020). For the selected CMAs and CPPs, a high-level
understanding of the process should be sought after through
a combination of studies assessing experimental design, sta-
tistical modelling, a mathematical modelling, and the under-
Table 2  (continued)

Capacity deviation
Quality Attributes

lying mechanisms.
When performing RA to identify and evaluate CMAs and
CPPs influencing CQAs, there are four steps: The first is
Endotoxin

Airtight
Volume
Aseptic

risk identification, in which risks are identified and listed.


The second step involved risk analysis performed using RA

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 659

Fig. 2  Compatibility test


between API and excipients

Table 3  Plackett–Burman Run API B C D E F G H I J K L M N


design for compatibility tests
1 + − + + − − − − + − + − + −
2 + + − + + − − − − + − + − +
3 + + + − + + − − − − + − + +
5 + − + + − + + − − − − + − −
6 + − − + + − + + − − − − + −
7 + + − − + + − + + − − − − +
8 + + + − − + + − + + − − − +
9 + + + + − − + + − + + − − +
10 + + + + + − − + + − + + − +
11 + − + + + + − − + + − + + −
12 + + − + + + + − − + + − + +
13 + − + − + + + + − − + + − −
14 + + − + − + + + + − − + + +
15 + − + − + − + + + + − − + −
16 + − − + − + − + + + + − − −
17 + − − − + − + − + + + + − −
18 + − − − − + − + − + + + + −
19 + + − − − − + − + − + + + +
20 + + + − − − − + − + − + + +

API active pharmaceutical ingredient, B Microcelac, C Starch, D microcrystalline cellulose (MCC), E


D-mannitol, F Lactose, G Crospovidone, H Sodium croscamellose (Na-CMC), I Sodium starch glycolate
(SSG), J Colloidal silicon dioxide, K Magnesium stearate (MG-St), L Sodium stearyl fumarate (SSF), M
citric acid, N Calcium hydroxide

tools, such as the preliminary hazard analysis (PHA). The Risk identification: first stage
third stage is risk evaluation, where major measures, such
as DoE, are taken through tools such as failure mode effects In risk identification, various material attributes (MAs) and
analysis (FMEA). As shown in Table 4, PHA might be use- process parameters (PPs) are listed using Ishikawa and fish-
ful when analysing existing systems or prioritising hazards bone diagrams, besides a process map. All potential vari-
where circumstances prevent a more extensive technique ables that can affect drug product quality are thus identified
from being used. Typically, hazards identified in the PHA through prior knowledge of the formulation and manufac-
are further assessed with other risk management tools such turing process (Figs. 3 and 4). After mapping the MAs and
as those in this section. Also, FMEA can prioritise risks and PPs that can affect CQA during the formulation step, risk
monitor the effectiveness of risk control activities. Finally, analyses should be performed.
the fourth stage is RA, with a goal to identify and organise When manufacturing a general immediate-release tab-
CMAs and CPPs affecting CQAs. let, or fixed-dose combination tablet, assuming the actual
process validation (PV) situation, risks may be incurred;

13
660

13
Table 4  Risk assessment tools
Tools Potential areas of use(s) References

Failure Mode Effects Analysis (FMEA) FMEA (see IEC 60812) provides for an evalu- FMEA can be used to prioritise risks and Guideline (2005), Sreedhara et al. (2015),
ation of potential failure modes for processes monitor the effectiveness of risk control Jayagopal and Murugesh (2020), Suresh et al.
and their likely effect on outcomes and/or activities (2015), (rights are reserved by Sharmada
product performance and Kakodkar (2015), Shah and Park (2009),
Failure Mode, Effects and Criticality Analysis FMECA can identify places where additional The output of an FMECA is a relative risk Jayagopal and Shivashankar (2017), Das et al.
(FMECA) preventive actions might be appropriate to “score” for each failure mode, which is used (2014), Chatterjee (2016), Oh et al. (2018)
minimize risks to rank the modes on a relative risk basis
Fault Tree Analysis (FTA) The FTA tool (see IEC 61025) is an approach FTA can be used to establish the pathway to
that assumes failure of the functionality of the root cause of the failure. FTA can be
a product or process. This tool evaluates used to investigate complaints or deviations
system (or sub-system) failures one at a time in order to fully understand their root cause
but can combine multiple causes of failure and to ensure that intended improvements
by identifying causal chains will fully resolve the issue and not lead to
other issues (i.e. solve one problem yet cause
a different problem)
Hazard Analysis and Critical Control Points HACCP is a systematic, proactive, and HACCP might be used to identify and manage
(HACCP) preventive tool for assuring product quality, risks associated with physical, chemical and
reliability, and safety (see WHO Technical biological hazards (including microbiologi-
Report Series No 908, 2003 Annex 7) cal contamination). HACCP is most useful
when product and process understanding is
sufficiently comprehensive to support identi-
fication of critical control points
Hazard Operability Analysis (HAZOP) HAZOP (see IEC 61882) is based on a theory HAZOP can be applied to manufacturing
that assumes that risk events are caused processes, including outsourced production
by deviations from the design or operating and formulation as well as the upstream
intentions suppliers, equipment and facilities for drug
substances and drug (medicinal) products
Preliminary Hazard Analysis (PHA) PHA is a tool of analysis based on applying PHA might be useful when analyzing existing
prior experience or knowledge of a hazard or systems or prioritising hazards where cir-
failure to identify future hazards, hazard- cumstances prevent a more extensive tech-
ous situations and events that might cause nique from being used. Typically, hazards
harm, as well as to estimate their probability identified in the PHA are further assessed
of occurrence for a given activity, facility, with other risk management tools such as
product or system those in this section
Journal of Pharmaceutical Investigation (2022) 52:649–682
Journal of Pharmaceutical Investigation (2022) 52:649–682 661

Fig. 3  Fishbone diagram (Ishikawa diagram)

Fig. 4  Mapping about MAs and PPs in the pharmaceutical development process

as such, all MAs and PPs required for process inspection sieving, post-mixing, tableting, and coating processes
and process control (IPC) must be listed. For example, gen- (Fig. 4), where presumably, a screening process is conducted
eral tablet manufacturing processes may include screening amongst each step. If the active pharmaceutical ingredient
(sifting), blending (dry mixing), wet granulation, drying, is aggregated, this material must be screened or sifted, after

13
662 Journal of Pharmaceutical Investigation (2022) 52:649–682

which the particle size distribution and flow ability of the P is defined as the frequency of a specific failure or event,
API powder will be MAs, and information on how to use the and it should refer to the P of the cause. Both P and S are
screen size, as well as mesh size are the PPs. In particular, scored along a scale of 1–4.
for the granulation process, impeller speed, chopper speed, The four point scale is based on the P of occurrence
amount of binder, etc. are specified as PPs. of failure is divided into (1) unlikely, (2) occasional, (3)
repeated, and (4) regular; whereas the scores based of the
Risk analysis and evaluation: second and third likely S effect on a drug product are divided into (1) minor,
stages (2) moderate, (3) major, and (4) extreme. The scale based
on the D of failure of detection is divided into (1) always
After listing and specifying all PPs, MAs, and QAs related detected, (2) regularly detected, (3) likely not detected, and
to the manufacturing, risk analysis begins. Broadly, in both (4) normally not detected. Failure risks were calculated
the second and third stages of RA, the tools PHA and FMEA using the risk priority number (RPN), which is the product
are used to determine the probability, severity, and detect- of P, D, and S (RPNs = P × D × S). The values of P, D, and S
ability based on a literature review and preliminary analy- for each failure mode tablets were assigned based on experi-
ses, ultimately ranking these variables based (Tables 5 and ence and literature review, and the RPNs were calculated.
6). Thereafter, the ripple effect of prioritised variables are A high RPN value for a concentration of disintegrant
evaluated via DoE or other experimental approaches, to indicates it could introduce a lag time in dissolution. The
improve process understanding and control strategies to be potential causes could be a low disintegrant concentration
developed (Shah et al. 2016; Casian et al. 2017; Patel et al. with a high occurrence value (P = 4), a low detectability
2017; Javed et al. 2018; Shekhawat and Pokharkar 2019). In value (D = 2), and an extreme severity value (S = 4); thus,
this risk analysis stage, RA tools are then used to perform a the calculated RPN for this failure mode was 32. Failure
simple analysis that enables the identification of materials modes with RPNs ≥ 30 were set as criteria for considering
and PPs affecting CQAs of the drug product. the DoE as a possible failure; however, RPN scores differ
In matrix analyses, such as PHA, CQA items can be by case. Further, many QbD papers have been identified that
related to identification, assay, uniformity, impurity, and use scales from 1 to 10. There is no clear standard or criteria
dissolution. Along the X-axis, the type of MAs used, or PPs for the values of P, S, and D.
applied in the drug production are listed (Tables 5a and 6a).
In Table 5a, if the amount of binding agent was increased, Quality risk assessment case of CMAs affecting CQAs
the dissolution profiles of a general tablet were released
slowly, and when the amount of binder was decreased, the The following is an example of the method to find CMAs
dissolution profiles indicated a rapid release; thus, the binder among various MAs affecting CQAs: API, binders, disinte-
amount affected dissolution profiles, potentially also affect- grants, lubricants, and stabilisers are used in the manufacture
ing bioavailability and efficacy of the finished drug product. of general tablets (Patel and Shelat 2017). However, because
PHA comprises a table in which each item is indicated the degree of risk depends upon the API, the RA must be
by a colour: green, yellow, or red corresponding to low-, made in advance during the development process. To assess
medium-, or high risk, respectively (Tables 5a and 6a). all cases that may occur during the manufacturing process,
Green indicates a broadly accepted level of risk, and no factor mapping should be performed. When MAs have been
further investigation is needed, whereas yellow indicates a successfully mapped, a literature search and initial screen-
degree of acceptable risk which may require further inves- ing studies for each factor of the composition variable are
tigation/justification to reduce the probability. Finally, red performed, and a PHA table can be established based on the
indicates an unacceptable risk that requires further investiga- results, classified into high (red), medium (orange), and low
tion for reduction. Accordingly, the amount of binder in the (green) according to the degree of risk (Table 5a). If PHA is
PHA table may be an unacceptable risk and is marked in red performed, FMEA are conducted to evaluate the initial risk
to indicate further research is needed. and criticality of each factor. At this time, S is scored from 1
The third stage of RA, risk evaluation, is performed to 4 points according to quality influence, P is scored accord-
through tools such as FMEA, to derive an equation that ing to the probability of occurrence, and D is scored accord-
enables the identification of MA and PP factors affecting ing to detectability. In addition, the RPN is calculated, with
the CQAs of the drug product. risk levels of 1–15, 16–30, and 31–64 classified into low,
Among the MAs, the functional groups of excipients and medium, and high risk, respectively. Physicochemical prop-
APIs are listed in the FMEA of Table 5b. For the corre- erties of API and excipients may affect the assay of the drug,
sponding analysis, each failure mode was ranked using an as well as content uniformity. In addition, there is a potential
estimated frequency of probability (P), the probability of an risk to the stability, dissolution pattern, and pharmacoki-
undetected failure later in the process (D), and severity (S). netic properties depending on the presence or absence of

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 663

Table 5  (a) Qualitative preliminary hazard analysis (PHA), and (b) quantitative failure mode effects analysis (FMEA) of material attributes
(MAs)

References Patricio et al. (2013), Sreedhara et al. (2015), Charoo et al. (2012), Jayagopal and Shivashankar (2017), Saydam (2016), Kovács
et al. (2021), Sharma et al. 2021, Jovanovska-Klincarska et al. (2018), Borde et al. (2016), Dias et al. (2021), Sitre and Kamble (2021), VS et al.
(2017)
CQAs critical quality attributes, CMAs critical material attributes, QTPP quality target product profile, API active pharmaceutical ingredient,
PSD particle size distribution, P probability, S severity, D detect ability, RPN risk priority number

specific excipients, which must be detected and managed were considered. Therefore, binders and disintegrants were
in advance. Using FMEA, when the RPN score was ≥ 30, it selected as CMAs. Glidant and lubricant posed moderate
was defined as CMA affecting CQAs (Table 5b). For MAs risks but were not CMA because they were deemed manage-
selected API, diluent, binder, granulating agent, disinte- able based on screening and pre-research data. If the amount
grant, wetting agent, glidant, anti-adherent, and lubricant of the binding solution selected as CMA is large, it may form

13
664 Journal of Pharmaceutical Investigation (2022) 52:649–682

Table 6  (a) Qualitative PHA, and (b) quantitative FMEA of process parameters (PPs)

References Bhattacharya et al. (2015), Nayak et al. (2017), Charoo et al. (2017), de la O Herrera et al. (2015), rights are reserved by Sharmada
and Kakodkar (2015), Jeyaprakash et al. (2020), Gupta and Khan (2013)
CQAs critical quality attributes, CPPs critical process parameters, QTPP quality target product profile, FBD fluidized bed dryer, CFM cubic feet
per minute, P probability, S severity, D detect ability, RPN risk priority number

hard granules and cause dissolution delay; whereas if the these factors were selected as CMAs, as they directly affect
amount is too small, an appropriate dissolution pattern can- the bioavailability of drugs, and must be optimised through
not be obtained. With a disintegrant, if too little is used, the additional experimental designs (Ha et al. 2018).
drug may not disintegrate within an appropriate time. Thus,

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 665

Quality risk assessment case of CPPs with CQAs against risk. For the overall RA, each selected CMA and
CPP were set as x values, and DoE was performed.
The following provides an example of the method to identify
CPPs among all PPs affecting CQA. The granulation process
is performed via fluidised bed granulation (FBG) and high Design of experiments (DoE) strategies
shear mixer equipment, and the raw material and binder are during drug development: selection
then added and kneaded. Because the risk depends on PP, of experimental design suitable
RA must be made in advance during the developmental pro- for the purpose
cess. To assess all possible cases that may occur during the
drug product manufacturing, one should similarly map the DoE is a data analysis method that helps plan, conduct, ana-
PPs. Once performed, a literature search and initial screen- lyse, and interpret controlled tests to determine the factors
ing studies of each composition variable can be conducted, affecting product quality, stability, or other key processes.
collectively informing the creation of a PHA table. PHA DoE helps identify all critical interactions by simultaneously
is similarly classified by colour according to the degree of manipulating multiple factors, furthermore, it supports the
risk, and FMEA is performed to evaluate the initial risk and robustness of the formulation and quality of the final product
criticality of each factor (Table 6a). At this time, P, D, and S by building experimental certainty and reproducibility into
are scored, and the RPN is calculated and classified into low, the process. Therefore, DoE must ensure that the QbD sys-
medium, or high risk. In the granulation process, the assay tem meets the design-specific quality requirements.
of the drug product may be affected due to API loss and However, an appropriate experimental design method
regarding the assay of granules. The physical properties of must be applied to the purpose of the experiment (Rathore
granules can affect product uniformity, and when the insolu- 2009). Scientific and systematic experimental design for
ble API is formed into amorphous granules by the process, drug development is a product of outputs in pursuit of a
there are potential risks to the dissolution pattern and phar- certain experimental purpose (i.e., designing an experiment
macokinetic properties; therefore, it must be detected and requires a purpose, and the appropriate research method to
managed in advance. For PPs, generally selected co-sifting, achieve said purpose) (Fig. 5) (Lee and Kim 2019). There-
blending, granulation, FBD, screening, lubrication, tablet- fore, to appropriately implement the DoE, the purpose of the
ing, and coating were considered (Table 6b). Also, when experiment must be defined with well-established hypoth-
the RPN score was ≥ 30, it was defined as CPP affecting the eses. The strategy of experimental design is divided into
CQAs. Therefore, granule manufacturing (impeller, chop- three categories: (1) Screening, (2) Optimisation, and (3)
per speed, and binder-granulating agent spraying rate), and Sequential experiments.
tableting (turret/feeder speed and compression/tamping pres- Screening experiments are efficient methods for identify-
sure) were selected as the CPPs. If the granulation process ing critical factors; however, the experimental design of the
time is long, large granules may be formed, which may harm screening experiment does not exist separately. Therefore, it
bioavailability and efficacy. According to tableting proper- is efficient to conduct a minimum experiment with many fac-
ties, granulation may affect the content uniformity of the tors. The experimental design most commonly used for this
tablet, capping, slug, etc., deleteriously affecting efficacy. purpose is the 2-level partial factorial design. Such screening
Therefore, the selected CPP should be optimised through the experiments are suitable for early stage or selection evalua-
additional experimental DoEs (Zandieh 2020). tion of new drugs in the feasibility test or RA stage.
The optimisation experimental design is suitable for
Risk assessment: fourth stage enhancing the response (Y) value by experimenting with
a few important factors. For optimisation, a curve response
Finally, when selecting CMAs among the MAs incorporat- model (typically a polynomial regression) should be cre-
ing API and excipients (Table 5b), the P, S, and D values of ated that can analyse the interaction between the independ-
FMEA are used to rank the score. If the RPN score is > 30 ent variable experimental factor, and the dependent variable
points (the hypothetical risk standard), it is considered a risk response factor in more detail; thus, experiments should be
that must employ measures such as DoE (i.e., it is selected designed to obtain higher-polynomial regression models.
as a CMA requiring DoE). Similarly, when selecting CPPs Optimisation experimental design and analysis methods
involved in the unit manufacturing process (Table 6), the include response surface methodology, and mixture experi-
results are also shown through PHA and FMEA, ultimately ments. In addition, the central composite and Box-Behnken
determining if the CPP requires DoE. In this way, binders, experimental designs are the primary methods for obtaining
disintegrants, granulation times, etc. were selected as CMAs the second-polynomial regression model. In drug composi-
and CPPs for which DoE's major measures should be taken tion research, simplex centroid, simplex lattice, and vertex

13
666 Journal of Pharmaceutical Investigation (2022) 52:649–682

Fig. 5  Road map for DoE selection in the pharmaceutical development process

experimental design methods are widely used for a mixture its development and establishment (Beg et al. 2015). For
of API and excipients. example, if a process is conducted near the boundary of the
Sequential experiments are an experimental strategy that design space, the risk of deviation from the design space
utilises both a screening experiment to find a major factor, may increase due to the normal process deviation (i.e., vari-
and an optimisation experiment to find an optimal response ation of general factors) (Fissore et al. 2015). Therefore,
value. This is most suitable for cases in which knowledge it is essential to verify the design space before performing
of research technology is lacking, or further exploratory scale-up for pharmaceutical manufacturing (Vo et al. 2020).
research is required. Designing an experiment with a sequen- Also, it is possible to improve the reliability of quality man-
tial experimental strategy is advantageous because optimisa- agement strategies for potential risks through the establish-
tion can be reached within a short time through systematic ment of control and operating space, besides design space
experiments. (Fig. 6) (Puskeiler et al. 2011). The control space is the area
of optimisation of process and formulation variables, and
Design space the operating space is defined in the manufacturing proce-
dure (Rathore 2009). After the design space has been estab-
Design space refers to a range designed to assure quality lished, if the process is operated within these limits, PV is
and identify complex interactions between MAs and PPs performed to demonstrate the provision of acceptable qual-
using DoE. The ICH has demonstrated that QA is provided ity (Rathore 2014). Through this, the control and operat-
by defining a design space as a multidimensional combina- ing space are established, and each set range is a drug that
tion and interaction of input and PPs (Raman et al. 2015). satisfies QTPP and CQA, with consistent quality within the
Thus, for CMAs and CPPs that directly affect CQAs selected range. In addition, it enables economic and efficient moni-
based on RA are parameters, interrelationships related to toring of drug quality, providing many advantages when
QA should be identified, as should multidimensional com- manufacturing and changing drug approvals. Consequently,
binations between variables. The design space representing running the processes within the design space is part of a
the multidimensional combinations ensures that a product quality control strategy (Peltonen 2018), and it is necessary
suitable for the established QTPP and CQA can be manu- to ensure that products suitable for QTPP and CQA can be
factured (Lee and Kim 2018). However, since drug develop- produced by deriving a design space related to the quality
ment creates a small-scale design space, an effective quality management strategy.
control strategy is needed to manage potential risks after

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 667

Fig. 6  Design, control, and normal operating space

Control strategy a uniformity test for unit amount is performed on the final
drug product rather than a content uniformity test in the
The control strategy is defined as ‘a planned set of con- compendium, a high level of quality assurance is possible,
trols, derived from current product and process under- along with the RTRT. The preferred control strategy technol-
standing, that assures process performance and product ogy is a real-time process analysis using process analytical
quality’, as described in ICH Q10 (Guideline 2008). The technology (PAT). This is an example of applying online and
main factors of the control strategy include material prop- in-line analysis methods to obtain qualitative or quantitative
erty management, process variable management, process information on intermediate and finished products (Miller
testing, intermediate material testing, procedure manage- 2020). Optical techniques and electrochemical methods
ment, final product management by release testing, sta- such as near-infrared spectroscopy, Raman analysis, laser
bility monitoring, and life cycle management (Karana- diffraction, and imaging are used for real-time monitoring
kov et al. 2012). In addition, when describing the control (Gavan et al. 2020). Assay, drying ratio, mixing ratio, tablet-
strategy for key factors, how the drug substance, semi- ing speed, and hardness of APIs and excipients in a general
finished product, container closure system, product, and tablet manufacturing process can be confirmed using Fourier
process inspection affect the final product quality must transform near-infrared (FT-NIR). In addition, the particle
be explained and justified (Ferreira and Tobyn 2015; de distribution, mixing ratio, granule, particle size, and mill-
Matas et al. 2016). These control factors should be based ing ratio of the API and excipient can be confirmed using
on product, formulation, and process understanding, and focused beam reflectance measurement (FBRM) (Murphy
should include at least CMA and CPP. Therefore, it is pos- et al. 2016). Thus, the control strategy in drug manufacturing
sible to manufacture while accounting for the variability of can guarantee consistent, high-quality products through test
the input drug substance through process understanding, management and process control (Singh et al. 2015).
and process management becomes possible to overcome
variability via adjustments to ensure consistent quality Lifecycle management
through knowledge surrounding this product and process
with quality risk management (Flåten et al. 2012; Markl Lifecycle management of pharmaceuticals is under discus-
et al. 2020). sion in ICH Q12, but commonly entails methods such as
As understanding of product performance increases, it define, measure, analyse, improve, control (DMAIC), design
is possible to judge property quality suitability via other for six sigma (DFSS), and product quality lifecycle imple-
methods for justification, namely the real-time release test- mentation (PQLI) based on PAT and in pursuit of under-
ing (RTRT) method. For example, a disintegration test can standing the process and evaluation technology of variables
be performed instead of a dissolution test with an immedi- and manufactured products (Jin et al. 2014). A scientific
ate-release tablet with a high API solubility. In addition, if approach must be applied to control and improve the quality

13
668 Journal of Pharmaceutical Investigation (2022) 52:649–682

throughout the entire process, from drug manufacturing to identification stage, all MAs and PPs were mapped through
delivery and patient consumption (von Stosch et al. 2016). a fishbone diagram until the final product was manufactured
One can confirm whether the process performance is main- and confirmed. As a wet granulation process was used, all
tained within the normal operating range (NOR) through factors in the formulation (solid dispersion granules, final
real-time monitoring (Ohage et al. 2016), and as part of mixture, and uncoated tablet, coating prescription), tab-
these monitoring activities, experience is accumulated let prescription, and coating prescription were mapped.
through routine manufacturing, permitting for trend analy- Potential CQA-impact risk was classified as high, medium,
sis throughout the manufacturing process. Further, regard- or low using the PHA tool. As QbD application involves
ing the design space using a mathematical model, periodic the detection and management of factors with potential
maintenance can help to verify and guarantee model perfor- risk (high and medium level) in advance, it was thus con-
mance (Rathore et al. 2017). Such model maintenance rep- firmed whether the selected factors affected the product’s
resents an example of an activity that can be managed under CQAs within the set range through initial screening studies.
its own quality system when the design space is held con- Such studies primarily used 2 k fractional factorial designs,
stant (Krummen 2015). When additional information about and based on the results, FMEA tools were used to evalu-
the process is obtained, the design space can be enlarged, ate each element in terms of estimated P, D, and S. The
reduced, and re-established as needed (Gouveia et al. 2018). corresponding RPN score was calculated, and CMAs and
CPPs that could affect CQAs were selected from among the
MAs and PPs (RPN > 30). When the amount of the binding
QbD drug development case study: solution increased, it had a critical effect on the dissolution
from QTPP to design space of the drug, affecting both the bioavailability and efficacy
(Table 9).
A case study presenting an example of QbD application to Because of performing RA on PPs in the same way, high
the wet granulation process commonly used in pharmaceuti- impeller and chopper speeds were found to potentially form
cal manufacturing was examined further. The corresponding large granules, possibly increasing tablet dissolution time.
paper. The primary research purpose was to develop tel- In addition, the longer the granulation time, the greater the
misartan potassium tablets via QbD that are the bioequiva- potential increase in dissolution time, as the larger granules
lent to the existing reference drug. Minitab® software (v.18, formed may affect bioavailability and efficacy; therefore, the
Minitab Inc., USA) was used for statistical analyses of the granulation time was selected as a CPP (Table 10).
experimental design, and an experimental method suitable As the curvature of the binding solution, disintegrant,
for each stage was selected. The initial QTPP step of specify- and granulation selected by CMA and CPP was confirmed
ing goals is listed in Table 7. Potential risks and feasibility in the screening experiment, a face-centred central com-
were described according to the viewpoint of pharmacists, posite design was applied. Central composite designs can
doctors, and patients. All items, save for the route of admin- efficiently estimate the first and second terms and model
istration, indications, and residual solvent, were selected as the curvature of response variables by adding experimen-
QTPP factors of risk. The considered CQAs were appear- tal points to the previously performed factorial design.
ance, identification, assay, dissolution, impurities, and resid- The input X value (factor) was three, and the level could
ual solvents directly related to the established QTPP. The be tested in a very narrow window of the knowledge space
risk associated with each item must be judged to determine through preliminary studies. The binder level was 4–10%,
whether establish a CQA, and the feasibility and basis for the disintegrant level was 2–6%, and the granulation time
this decision must be presented (Table 8). Product dissolu- was 3–7 min. The table obtained through the design is listed
tion and impurities can affect bioavailability and stability; in Table 10, and the total number of experiments was n = 20
thus, both were selected as CQAs. Sometimes, it is possible ­(23 + (2 × 3) + 6). Finally, the output Y (response) value was
to confirm whether one can substitute for other quality char- selected as hardness (kp), friability (%), disintegration (min),
acteristics, and whether there can be variability in output and drug dissolution (%) regarding QTPP and CQA. After
variables. Also, if it is determined there will be changes completing all experiments, results for disintegration time,
due to the composition or variation of manufacturing PPs, hardness, friability, dissolution, impurities were input to the
it should be set as an output variable. statistical program, and model lack of fit tests were checked
RA is conducted after QTPP and CQAs selection, then through design analysis. Here, if p > 0.05, it was judged
compatibility test are performed. All parameters, such as that the null hypothesis could not be rejected for lack of fit.
composition, mixing, granulation, lubrication, and filling Therefore, after DoE modelling was optimised, the results
processes should be detected in advance, as they can affect could be interpreted through programs such as Pareto charts
CQA. RA was performed to select and evaluate CMAs and and residual plots for each response value (Y 1–5).
CPPs, the primary factors influencing CQAs. In the risk

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 669

Table 7  Quality target product


profile (QTPP; reproduced with
permission of Oh et al., Drug
Development and Industrial
Pharmacy, Taylor & Francis
Online, 2018)

13
670 Journal of Pharmaceutical Investigation (2022) 52:649–682

Table 8  Critical quality


attributes (CQAs; reproduced
with permission of Oh et al.,
Drug Development and
Industrial Pharmacy, Taylor &
Francis Online, 2018)

Modelling can be optimised by selecting significant was systematically established with all individual receptive
factors through the Pareto chart (Fig. 7a), and the statis- areas of each CMA and CPP.
tical significance can be confirmed through residual plots Telmisartan potassium tablet was manufactured by the
(Fig. 7b). In addition, the degree of effect on the output wet granulation method using a high shear mixer, accord-
can be checked through the slopes of the main effects and ing to the formulation composition in the design space.
interaction plots (Fig. 7c, d). Finally, the numerical effect The test and reference drug (Micardis®), which are the
of each element on the output can be accurately obtained final composition within the design space, were compared
through the contour and surface diagrams (Fig. 7e, f). As through the pharmacokinetic profile of the beagle dog,
shown in the contour plot, the hardness increased with the and the results are shown in Fig. 8. Consequently, the 90%
amount of binder, whereas the amount of disintegrant had confidence interval (90% CI 0.9071–1.0814) for the geo-
little effect. By overlapping the contour plots based on these metric least-squares mean ratio of the area under the curve
statistical results, it is possible to obtain a superimposed plot (AUC) was 0.9790 for the point estimate. In addition, the
capable of confirming the optimal design value of the true bioequivalence criterion for telmisartan was within the
space being operated. The target values for the individual range of 0.80–1.25; therefore, through pre-clinical stud-
responses of the formulations are shown in Fig. 7, in which ies, the test drug developed via the application of QbD,
it is possible to set a response value region where all criteria and the reference drug were proven to be bioequivalent.
are satisfied (i.e., the design space area can be confirmed). By applying QbD, risks were detected and managed in
As the model was developed and validated, the design space advance, improving product quality to a high level, besides

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 671

Table 9  (a) Qualitative preliminary hazard analysis (PHA), and (b) quantitative failure mode effects analysis (FMEA) of material attributes
(MAs; reproduced with permission of Oh et al., Drug Development and Industrial Pharmacy, Taylor & Francis Online, 2018)

13
672 Journal of Pharmaceutical Investigation (2022) 52:649–682

Table 10  (a) Qualitative preliminary hazard analysis (PHA), and (b) quantitative failure mode effects analysis (FMEA) of process parameters
(PPs; reproduced with permission of Oh et al., Drug Development and Industrial Pharmacy, Taylor & Francis Online, 2018)

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 673

Fig. 7  Identification of CMA and CPPs using: a Pareto charts and b space using face-centered central composite design (reproduced with
residual plots; Effects of CMAs and CPPs using: c main effects plot, permission of Oh et al., Drug Development and Industrial Pharmacy,
d interaction plot, e contour plot, f response surface plot; g Design Taylor & Francis Online, 2018)

Fig. 7  (continued)

13
674 Journal of Pharmaceutical Investigation (2022) 52:649–682

Fig. 8  Plasma concentration–


time profiles of Micardis®
and TP tablets at an 80 mg
dose telmisartan base in
beagle dogs. Data represent
the mean ± standard devia-
tion (SD, n = 12). Reference
tablet, Micardis® (telmisartan
free base tablet); TP tablet,
telmisartan potassium tablet
(reproduced with permission of
Oh et al., Drug Development
and Industrial Pharmacy, Taylor
& Francis Online, 2018)

the manufacturing process to increase efficiency. Ulti- the overlapping contour plots, and the DS of the response
mately, this study is expected to reduce costs by minimis- variable and influencing factor established the criteria veri-
ing unnecessary operation of equipment throughout the fied within the 95% confidence level. The final composi-
process. tion of the dry-coated tablet was determined, and a process
study was conducted to identify the QTPP. The optimised
formulation was physicochemically stable for > 24 months,
QbD application case studies and in a pharmacokinetic study of beagle dogs, the ­Tmax was
in the pharmaceutical development stage approximately thrice as fast as the reference drug of rabe-
prazole (Pariet®). Formulation development applying QbD
Development of immediate‑release rabeprazole guaranteed the quality of the final drug because it detected
sodium dry‑coated tablet and managed various risks in advance. With rabeprazole
sodium used as the API here, effective quality control was
Lee and Kim (2021) conducted a study on the development possible when comparing the stability of the formulation
of rabeprazole sodium and sodium bicarbonate fixed-dose to that of the existing reference drug with poor stability.
combination tablets through the QbD approach. The purpose Therefore, the QbD system strategy provided information
was to develop a treatment for gastroesophageal reflux dis- for drug development, which involved sensitive issues in
ease that, unlike the existing PPI formulations, can be rap- drug manufacturing, such as stability and tableting proper-
idly absorbed by the stomach, applying the QbD approach ties (Lee and Kim 2021).
to the development process. Parameters of QTPP included
clinical use, route of administration, formulation, delivery Development of topical hydrogel containing
system, content, container, and packaging, API release or ketoconazole loaded cubosomes
delivery, characteristics affecting pharmacokinetic proper-
ties, sterility, purity, stability, and dissolution. The QTPPs Rapalli et al. (2021) explored the development of topical
were evaluated for feasibility, and to establish the direction hydrogel containing ketoconazole loaded cubosomes with
of product and process development, the CQAs were iden- lower surfactant concentrations via the QbD approach. The
tified through them and prior knowledge. To identify the secondary step in the QbD design was identification and
CQAs, physical characteristics, appearance, identification, selection of CQAs. As CQAs are the prominent product QA
dissolution, impurities, weight variation, content uniform- selected from the QTPP which influence the finished dos-
ity, assay, and residual solvents were specified. For the RA, age form, their effects required exploration and monitoring.
PHA and FMEA were used based on prior knowledge and Surfactant and stabiliser concentration were selected as the
initial experimental data. An RPN > 30 was defined as a dominant MAs affecting particle size and size distribution,
CMA or CPP. The central composite design of DoE was respectively. These two properties affect entrapment effi-
used to establish an appropriate management strategy for ciency and drug release (%). Stirring speed and stirring time
the CQAs of the outer layer and optimise its binding and were selected as CPPs because they could affect the size
disintegration properties. The design space was derived from of the pre-emulsion and entrapment efficiency by avoiding

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 675

coalescence of the two oil types. High RPN scores ≥ 100 penetration. When manufacturing pharmaceuticals, critical
were critical parameters/attributes. Screening and optimi- factors can function as various variables in the final prod-
sation of formulations with 32 factorial designs were con- uct; therefore, it is imperative to investigate the interaction
ducted with Design-Expert® (version 9.0, Stat-Ease Inc.). of critical factors through minimal experimentation using
Keeping the combination of constituents similar to that of DoE. This study provided knowledge of the more efficient
the optimised batches, as predicted post DoE analyses, scale- application of QbD by describing its use topical formulation
up batches were prepared. The 32 factorial design model development (Patel et al. 2021).
successfully predicted the composition of the optimised
formulation within the confidence limits, with the produced Development of liposomes including hydrophilic
ketoconazole loaded cubosomes showing a release pattern APIs
similar to the Korsmeyer–Peppas model experiencing Fick-
ian diffusion (67% cumulative ketoconazole release within Xu et al. (2011) is a paper on the development of liposomes
24 h). Ex vivo permeation study of hydrogel containing including hydrophilic APIs also applied the QbD approach.
ketoconazole loaded cubosomes revealed a sustained release Tenofovir was selected as the hydrophilic API, and the
pattern through goat ear skin with ~ 92.73% release within 24 potential risk variables were selected through RA, before
h. Thus, with drug development addressing the problem of the Plackett–Burman design was used to confirm the effect
solubility, using surfactants was necessary; however, exces- of liposomes on the properties (drug encapsulation effi-
sive use of surfactants in drug development can affect patient ciency, particle size, and physical stability). Accordingly,
safety. Overall, it is judged that the QbD system strategy it was confirmed that lipid and drug concentration had a
here was appropriately utilised in formulations for develop- major effect on drug encapsulation. The multidimensional
ing ribosomes as a method of improving solubility (Rapalli interaction between lipid concentration, drug concentration,
et al. 2021). and encapsulation efficiency was investigated through cen-
tral composite design, and design space was also derived.
Development of topical arginine solid lipid Thus, a liposome formulation containing hydrophilic APIs
nanoparticles was optimised via the QbD approach. This helped to ensure
product quality by minimising variability (Xu et al. 2011).
Patel et al. (2021) assessed development through QbD for Plackett–Burman design is efficient at identifying critical
additional rational forethought and step-by-step develop- factors in the experimental design, and the interactions
ment of solid lipid nanoparticles (SLNs). Considering the between various factors can be confirmed through minimal
type of dosage form and formulation method, the QTPP was experimentation. Therefore, the application of the QbD sys-
established, and a list for SLNs can be found in this paper. tem using the Plackett–Burman design effectively informed
For CQA identification, the Ishikawa diagram was devel- critical factor selection and optimisation.
oped, providing a transparent picture of CPPs and CMAs
affecting the particular outcomes of each variable. FMEA Liposomes for nasal administration
was established to classify the parameters with higher risks
of failure in the quality of SLNs, and the CPPs and CMAs Pallagi et al. (2019) assessed the early development of
with the highest RPN were the concentrations of surfactant, liposomes for nasal administration through the QbD
oleic acid, and the sonication amplitude. Following FMEA approach. In this study, the authors adapted QbD and RA
analysis (RPN number) and preliminary screening studies, methods to the early development phase of a new nano-
the three highest ranked CPs that influenced the final formu- sized liposomal formulation for nasal administration with
lation quality were included in DoE by applying 3 factor-2 brain target. This research displayed how to apply this risk-
level experimental design for SLN optimisation. Therefore, focused approach concentrated on the first four stages of
by utilisation of all the concepts, the SLNs containing a con- QbD implementation. In this way the QTPP was defined, the
centration of 2.00% poloxamer 188, 1.5% oleic acid, and critical factors were identified, and an RA was performed.
59.78% sonication amplitude was optimal, having a particle The RA results helped in the factorial design-based lipo-
size of 273.6 nm, and 8.57% of drug loading. This research some preparation by the lipid film hydration method. The
revealed the significance of the QbD approach in develop- initial step was to define the TPP and its quality criteria,
ing a novel drug delivery system with fewer experimental where diagrams are useful for QTTP selection, as well as
trials by applying the predicted model of DoE software. As the critical factors during the next QbD-guided develop-
per the concept of QbD, the FMEA method combined with ment steps. The second step was to select the factors with
the experimental design was appropriate for optimising the critical effects on the targeted product quality. CQAs may
critical parameters affecting QTPP. With topical formula- include information on particle or vesicle size, size distri-
tions, particle size and drug loading affected QTPP for skin bution, drug release, etc. In the next step, the CMAs and/

13
676 Journal of Pharmaceutical Investigation (2022) 52:649–682

or the CPPs potentially affecting CQAs were determined. further evaluation. DXIBN transdermal patches were for-
In the RA-based formulation development, the critical fac- mulated using various concentrations of selected poly-
tors were analysed, the interrelations between them were meric excipients (matrix material: ethyl cellulose and pol-
estimated, and the factors were ranked by their critical effect yvinylpyrrolidone, plasticiser (di-N-butyl phthalate) and
on the final product. The RA results helped establish the a conventional permeation enhancer (almond oil). Initial
experimental design. The Main Effect Screening Design 2 patch formulations were evaluated for their physiochemi-
program was selected for screening the effects of the for- cal properties (such as the thickness, moisture uptake,
mulation parameters on the quality of the final liposomal final moisture content, and DXIBN content). Also, impact
product and revealed the importance of the concentration of patch components on resulting tensile strength and
of phospholipids (Phospholipon 90G®), which were the in vitro permeation were used to predict an optimal patch
third data on the list of the CQAs and chosen as variable formulation using a QbD approach, which was evaluated
X1. According to the selection methodology, the size of the and compared to a commercial oral tablet dosage form
liposome vesicles and size distribution, located in the first for in vitro and in vivo release (rabbit model). Predicted
position on the critical list of CQAs, were selected as the properties (tensile strength and DXIBN steady state flux)
response variable and second factor, respectively, whereas for the QbD optimised formulation closely agreed with
the API was held constant. From the CPPs, the temperature experimental results, and the QbD optimal patch formula-
of ultrasonication was chosen as variable X2, and because tion behaviour differed significantly from the commercial
the type of the organic solvent was constant, the critical fac- tablet formulation in vivo. Such QbD approach model
tor pertaining to the number of the second filtration was based predictions thus reduced cost and time in formu-
designated as variable X3. The investigational results of the lation development sciences here, besides the derived
prepared liposomes (API and API-free samples were pre- formulation maintaining a shorter onset time than the
pared following the same factorial design pattern), namely previously developed oral tablet (Propen®, Global Phar-
vesicle size, size distribution, specific surface area, and sur- maceuticals, Pakistan), with long-lasting effects. Consid-
face characteristics, verified the exactness of the RA and ering the scale-up step, applying QbD can be considered
the critical factor-based theoretical prediction showed strong an industrially useful strategy for minimising cost and
relationships between the product design (composition of the time (Akhlaq et al. 2016).
liposomes and PPs, such as temperature, number of filtra-
tions, etc.) and the product characteristics of the prepared Development of resveratrol‑loaded mucoadhesive
liposomes. The results demonstrated how the QbD approach lecithin/chitosan nanoparticles for prolonged ocular
could improve the formulation process in the development drug delivery
of liposomes, lead to an effective product preparation pro-
cess, and help in the optimisation and the rationalisation Saha et al. reported on drug-containing nanoparticles
of liposomal development, even in special circumstances using the QbD approach. The QbD workflow began with
when a lipophilic active ingredient was incorporated into defining the patient-centric QTPPs, considering the qual-
the liposomes. This research contained explanations of the ity, safety‚ and efficacy of the product. Then, the relative
development of liposomes with the QbD approach applied potential QAs were identified, and their appropriate targets
to drugs with low solubility. It was judged that through this were fixed with respect physicochemical and biological
innovative design and development model, it was possible properties of the intended lecithin/chitosan mucoadhesive
to provide a platform to help resolve problems of drugs with nanoparticles based on the literature and experimental
low solubility (Pallagi et al. 2019). values. Next, the MAs, PPs, and other factors likely to
affect the QAs, were illustrated by the fishbone diagram as
Development of a transdermal patch the initial RA, and the high- and medium-risk QAs were
considered as CQAs. Ten CQAs obtained from the RA
Akhlaq et al. pursued the development of a transdermal study were experimented upon by the definitive screening
patch for dexibuprofen (DXIBN) using the QbD approach. design (DSD) to determine the influential CPPs and CMAs
To optimise patch formulation, Box–Behnken (three-fac- for further optimisation, DoE analysis. The final RA of
tor-three-level) design was used for the determination of the MAs and PPs were performed in two steps: The high-
quadratic response surfaces with Design-Expert®. The and medium-risk MAs and PPs were considered CMAs
polynomial equation (Eq. 2) was generated, and the mod- and CPPs, respectively. Following the determination of
elled response was validated using the F-value (ANOVA; two crucial CMAs from DSD, a two-factor, five-level,
significant terms identified as values of ‘Prob ˃ F’ < 0.05. eleven run central composite design (CCD) was gener-
The modelled steady state flux of selected patches was ated to optimise and define a design space. For the CCD,
thereafter experimentally obtained and subjected to the lecithin concentration and drug concentrations were

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 677

expressed by X1 and X2, respectively. Thus, the experi- Conclusion


ments were planned based on DoE; accordingly, the design
point with the highest desirability value provided a nano- In the pharmaceutical industry, drug development is lim-
sized, stable, and optimum drug-entrapped formulation. ited by product development and quality, as these essential
The optimised formulation, developed within the lab scale, steps rely on experience. These issues can be overcome by
met the pre-defined, patient-centric QTPPs targets. This introducing a QbD system, which utilizes science and statis-
study provided a case of successful development of QbD- tics-based technologies. Applying QbD based on risk man-
based resveratrol-loaded mucoadhesive lecithin/chitosan agement enables the development of high-quality and safe
nanoparticles for prolonged ocular drug delivery; however, drugs for patients. This review paper is expected to solve
it has limitations, as it was developed and optimised in the difficulties surrounding drug development via QbD by
a laboratory-scale. Therefore, it is necessary to set the providing accurate information and application methods for
applicable range in the industry within the design space, a functional system.
set through a scale-up study along with the experimental
design (Saha et al. 2021).
Funding This research was supported by the Industrial Strategic Tech-
Biopharmaceutical nology Development Program (20008909, Development of AI-based
smart platform for pharmaceutical formulation design and manufac-
turing process) funded by the Ministry of Trade, Industry & Energy
Sharma et al. (2021) analysed the improvement of the (MOTIE, Korea).
biological properties of sorafenib tosylate (SFN) through
QbD. To improve the biological properties of SFN, a type Declarations
III self-emulsifying delivery system (Type III-SEDDS)
was applied, with an optimised formulation through the Conflict of interest All authors (S.‑H. Lee, J.‑K. Kim, J.‑P. Jee, D.‑J.
Jang, Y.‑J. Park, and J.‑E. Kim) declare that they have no conflict of
QbD approach. Initially, the QTPP embarked upon defin- interest.
ing all the requisite quality characteristics of the formula-
tion to be developed for achieving maximal therapeutic Research involving human and animal rights This article contains no
efficacy of SFN, and CQAs were selected. Through first- studies with human and animal subjects performed by the authors.
order 7-variables-2-level Taguchi OA design, with eight
experimental runs, screening tests were performed for
MAs and PPs that could create risk for the CQA. Based on
the results of preformulation and screening studies, type References
III LFCS SEDDS formulations were optimised, employ-
ing a 3-factor-3-level D-optimal mixture design. A total Abhinandana P, Nadendla R. Application of quality by design and its
parameters for pharmaceutical products
of 16 Type III-SEDDS formulations, including the quin- Akhlaq M, Arshad MS, Mudassir AM, Hussain A et al (2016) For-
tuplicate runs, were prepared according to the D-optimal mulation and evaluation of anti-rheumatic dexibuprofen trans-
design matrix and characterised for CQAs: % drug release dermal patches: a quality-by-design approach. J Drug Target
at 60 min (Rel60), Dnm, mean dissolution time (MDT) and 24(7):603–612
Aksu B, Mesut B (2015) Quality by design (QbD) for pharmaceu-
% dissolution efficiency (DE). The optimised formulation tical area. İstanbul Üniversitesi Eczacılık Fakültesi Dergisi
improved the dissolution rate by ~ 8 times or more, whereas 45(2):233–251
the drug permeability and absorption also increased > 8 Alt N, Zhang TY, Motchnik P, Taticek R et al (2016) Determination of
times compared to the conventional SFN. Therefore, using critical quality attributes for monoclonal antibodies using quality
by design principles. Biologicals 44(5):291–305
QbD optimised the formulation, and the bioavailability of Amasya G, Aksu B, Badilli U, Onay-Besikci A et al (2019) QbD
SFN was improved. Also, stability studies of SFN formu- guided early pharmaceutical development study: production of
lations indicated that the ambient storage conditions (i.e., lipid nanoparticles by high pressure homogenization for skin
25 °C) were suitable for maintaining the robustness of the cancer treatment. Int J Pharm 563:110–121
Anderson A, McGibbon GA, Bhal SK (2017) Achieve QbD by manag-
self-emulsifying formulation, reporting inconsequential ing impurity data: cheminformatics enables QbD strategies for
changes in the CQAs. (Sharma et al. 2020). Compared process and analytical impurity control. Genet Eng Biotechnol
to synthetic drugs, biopharmaceuticals have many criti- News 37(20):26–27
cal factors sensitive in scope, so it is necessary to detect Arora D, Khurana B, Narang R, Nanda S (2016) Quality by design
(QbD) approach for optimization and development of nano drug
and manage risks in advance; therefore, through this study, delivery systems. Trends Drug Deliv 3:23–32
an appropriate QbD system strategy for biopharmaceuti- Ban E, Jang D-J, Kwon T-H, Kim A (2019) Pharmaceutical
cal development was identified. Furthermore, an efficient GMP inspection: case study of USA and EU. Yakhak Hoeji
description of the Taguchi OA design and D-optimal mix- 63(1):15–23
ture design was also put forth.

13
678 Journal of Pharmaceutical Investigation (2022) 52:649–682

Beg S, Sandhu PS, Batra RS, Khurana RS et al (2015) QbD-based Colobatiu L, Gavan A, Mocan A, Bogdan C et al (2019) Development
systematic development of novel optimized solid self-nanoe- of bioactive compounds-loaded chitosan films by using a QbD
mulsifying drug delivery systems (SNEDDS) of lovastatin approach–a novel and potential wound dressing material. React
with enhanced biopharmaceutical performance. Drug Deliv Funct Polym 138:46–54
22(6):765–784 Cunha S, Costa CP, Loureiro JA, Alves J et al (2020a) Double opti-
Beg S, Hasnain MS, Rahman M, Swain S (2019) Introduction to qual- mization of rivastigmine-loaded nanostructured lipid carriers
ity by design (QbD): fundamentals, principles, and applications. (NLC) for nose-to-brain delivery using the quality by design
Pharmaceutical quality by design. Elsevier, pp 1–17 (QbD) approach: formulation variables and instrumental param-
Bhambure R, Kumar K, Rathore AS (2011) High-throughput process eters. Pharmaceutics 12(7):599
development for biopharmaceutical drug substances. Trends Cunha S, Costa CP, Moreira JN, Lobo JS et al (2020b) Using the
Biotechnol 29(3):127–135 quality by design (QbD) approach to optimize formulations of
Bhattacharya J, Pharm M, Phil M (2015) Quality Risk Management- lipid nanoparticles and nanoemulsions: a review. Nanomedicine
Understanding and control the risk in pharmaceutical manufac- 28:102206
turing industry. Int J Pharm Sci Invent 4(1):29–41 Dangre PV, Phad RD, Surana SJ, Chalikwar SS (2019) Quality by
Bhoop BS (2014) Quality by Design (QbD) for holistic pharma excel- design (QbD) assisted fabrication of fast dissolving buccal film
lence and regulatory compliance. Pharm times 46(8):26–33 for clonidine hydrochloride: exploring the quality attributes. Adv
Bhusnure O, Nandgave A, Gholve SB, Thonte SS et al (2015) Formu- Polym Technol 2019:1–13
lation and evaluation of fast dissolving tablet on montelukast Das A, Kadwey P, Mishra JK, Moorkoth S (2014) Quality Risk Man-
sodium by using QbD approach. Indo Am J Pharm Sci 5:1092 agement (QRM) in pharmaceutical industry: tools and methodol-
Bhutani H, Kurmi M, Singh S, Beg S et al (2004) Quality by design ogy. Int J Pharm Qual Assur 5(3):13–21
(QbD) in analytical sciences: an overview. Qual Assur 3:39 Desai S (2018) Quality by Design-based formulation and evaluation of
Borchert D, Suarez-Zuluaga DA, Thomassen YE, Herwig C (2020) fast dissolving tablet of aspirin. Asian J Pharm 12(01)
Risk assessment and integrated process modeling–an improved DESIGN Q.B (2012) Quality by Design (QbD) of sterile dosage form
QbD approach for the development of the bioprocess control packaging
strategy. AIMS Bioeng 7(4):254–271 de la Herrera MA, Luna AS, da Costa ACA, Lemes EMB (2015) A
Borde SD, Shahi SR, Kale KV, Jadhav AC et al (2016) Formulation structural approach to the HAZOP–Hazard and operability tech-
and evaluation of Paliperidone HCl mouth dissolving tablet by nique in the biopharmaceutical industry. J Loss Prev Process
QbD approach. Pharm Biol Eval 3(6):528–540 Ind 35:1–11
Braem A, Turner G (2019) Applications of quality risk assessment de Matas M, De Beer T, Folestad S, Ketolainen J et al (2016) Strate-
in Quality by Design (QbD) drug substance process develop- gic framework for education and training in Quality by Design
ment. In: Chemical engineering in the pharmaceutical industry: (QbD) and process analytical technology (PAT). Eur J Pharm
active pharmaceutical ingredients, pp 1073–1089 Sci 90:2–7
Calnan N, O’Donnell K, Greene A (2013) Enabling ICH Q10 imple- Deng Y, Zhong G, Wang Y, Wang N et al (2019) Quality by design
mentation—Part 1 Striving for excellence by embracing ICH approach for the preparation of Fat-soluble vitamins lipid inject-
Q8 and ICH Q9. PDA J Pharm Sci Technol 67(6):581–600 able emulsion. Int J Pharm 571:118717
Casian T, Iurian S, Bogdan C, Rus L et al (2017) QbD for pedi- Dias AL, Ferreira NN, Ferreira LMB, Pedreiro LN et al (2021) Regula-
atric oral lyophilisates development: risk assessment fol- tion of nanotechnology-based products subject to health regula-
lowed by screening and optimization. Drug Dev Ind Pharm tions: application of Quality by Design (QbD) and Quality Risk
43(12):1932–1944 Management (QRM). In: Nanocarriers for drug delivery: con-
Charoo NA, Shamsher AA, Zidan AS, Rahman Z (2012) Quality by cepts and applications, pp 319–347
design approach for formulation development: a case study of Dispas A, Avohou HT, Lebrun P, Hubert P et al (2018) ‘Quality by
dispersible tablets. Int J Pharm 423(2):167–178 Design’ approach for the analysis of impurities in pharmaceuti-
Charoo NA, Cristofoletti R, Kim SK (2017) Integrating biopharmaceu- cal drug products and drug substances. TrAC Trends Anal Chem
tics risk assessment and in vivo absorption model in formulation 101:24–33
development of BCS class I drug using the QbD approach. Drug Djuris J, Ibric S, Djuric Z (2013) Quality-by-design in pharmaceutical
Dev Ind Pharm 43(4):668–677 development. Computer-aided applications in pharmaceutical
Chatterjee B (2016) Applying lean six sigma in the pharmaceutical technology. Elsevier, pp 1–16
industry. Routledge, London Dutta J, Samanta A, Dey S, Andhale G et al. Quality by Design: a
Chaudhury A, Barrasso D, Pandey P, Wu H et al (2014) Population bal- revolutionary concept of pharmaceutical product development
ance model development, validation, and prediction of CQAs of a in modern era
high-shear wet granulation process: towards QbD in drug product Ferreira AP, Tobyn M (2015) Multivariate analysis in the pharmaceuti-
pharmaceutical manufacturing. J Pharm Innov 9(1):53–64 cal industry: enabling process understanding and improvement
Chavan SD, Pimpodkar NV, Kadam AS, Gaikwad PS (2016) Quality in the PAT and QbD era. Pharm Dev Technol 20(5):513–527
by design. Asian J Res Pharm Sci 6(1):45–50 Finkler C, Krummen L (2016) Introduction to the application of QbD
Chavda H (2016) Qbd in developing topical dosage forms. Ely J Pharm principles for the development of monoclonal antibodies. Bio-
Res 2(1):1–2 logicals 44(5):282–290
Chen C (2006) Implementation of ICH Q8 and QbD—an FDA perspec- Fissore D, Pisano R, Barresi AA (2015) Using mathematical mod-
tive. PharmaForum Yokohama, June eling and prior knowledge for QbD in freeze-drying processes. In
Chordiya MA, Gangurde HH, Sancheti VN (2019) Quality by design: Quality by design for biopharmaceutical drug product develop-
a roadmap for quality pharmaceutical products. J Rep Pharm ment. Springer, pp 565–593
Sci 8(2):289 Flåten GR, Ferreira AP, Bellamy L, Frake P (2012) PAT within the
Chudiwal VS, Shahi S, Chudiwal S (2017) Article details develop- QbD framework: real-time end point detection for powder blends
ment of sustained release gastro-retentive tablet formulation of in a compliant environment. J Pharm Innov 7(1):38–45
bepotastine besilate using Quality by Design (QbD) approach. Freeman T, Birkmire A, Armstrong B (2015) A QbD approach to con-
Indian Drugs 54:48–60 tinuous tablet manufacture. Procedia Eng 102:443–449

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 679

Fukuda IM, Pinto CFF, Moreira CS, Saviano AM et al (2018) Design estimate nintedanib degradation products and their pathways.
of experiments (DoE) applied to pharmaceutical and analytical Arab J Chem 13(9):7087–7103
quality by design (QbD). Brazil J Pharm Sci. https://​doi.​org/​10.​ Jeyaprakash RM, Sharma A, Bora R, Chandra A (2020) Impact of qual-
1590/​s2175-​97902​01800​00010​06 ity risk management process in pharmaceutical industry to curtail
Gandhi A, Roy C (2016) Quality by design (QbD) in pharmaceuti- the non-conformity. Int J Pharm Qual Assur 11(01):179–185
cal industry: tools, perspectives and challenges. PharmaTutor Jin S-E, Kim M-S, Shin S, Jeong SH et al (2014) Current status and
4(11):12–20 understanding of QbD implementation in pharmaceutical indus-
Gavan A, Porfire A, Marina C, Tomuta I (2017a) Formulation and phar- try. Korean Soc Food Drug Cosmet Regul Sci 9(2):141–148
maceutical development of quetiapine fumarate sustained release Jovanovska VP (2018) Development and formulation optimisation
matrix tablets using a QbD approach. Acta Pharm 67(1):53–70 of modified release dosage form using quality by design–QbD
Gavan A, Porfire A, Marina C, Tomuta I (2017b) Formulation and aproach
pharmaceutical development of quetiapine fumarate sustained Jovanovska-Klincarska I, Popovska I, Kostovski V, Dimitrovska J
release matrix tablets using a QbD approach. Acta Pharm et al (2018) Risk assessment in pharmaceutical microbiology.
67(1):53 J Hyg Eng Des 25:7–13
Gavan A, Iurian S, Casian T, Porfire A et al (2020) Fluidised bed gran- Kadam VR, Patil M, Pawar VV, Kshirsagar S (2017) Quality by
ulation of two APIs: QbD approach and development of a NIR Design (QbD). Asian J Res Pharm Sci 7(4):197
in-line monitoring method. Asian J Pharm Sci 15(4):506–517 Kader M (2016) Mitigating the risks of generic drug product devel-
Gouveia FF, Felizardo PM, Menezes JC (2018) Lifecycle management opment: an application of Quality by Design (QbD) and Ques-
of PAT procedures: applications to batch and continuous pro- tion based Review (QbR) approaches. J Excip Food Chem
cesses. In: Multivariate analysis in the pharmaceutical industry. 7(2):915
Elsevier, pp 323–345. Kajiwara E, Kamizato H, Shikano M (2020) Impact of Quality
Grangeia HB, Silva C, Simões SP, Reis SP (2020) Quality by design in by Design development on the review period of new drug
pharmaceutical manufacturing: A systematic review of current approval and product quality in Japan. Ther Innov Regul Sci
status, challenges and future perspectives. Eur J Pharm Biopharm 54:1192–1198
147:19–37 Kan S, Lu J, Liu J, Wang J et al (2014) A quality by design (QbD)
Guideline I.H.T (2005) Quality risk management. Q9, Current step. case study on enteric-coated pellets: screening of critical vari-
4. 408 ables and establishment of design space at laboratory scale.
Guideline I.H.T (2008) Pharmaceutical quality system q10. Current Asian J Pharm Sci 9(5):268–278
Step 4 Kanwal U, Mukhtar S, Waheed M, Mehreen A et al (2021) Fixed
Gupta S, Jhawat V (2017) Quality by design (QbD) approach of phar- dose single tablet formulation with differential release of
macogenomics in drug designing and formulation develop- amlodipine besylate and simvastatin and its pharmacokinetic
ment for optimization of drug delivery systems. J Control Rel profile: QbD and risk assessment approach. Drug Des Dev
245:15–26 Ther 15:2193
Gupta A, Khan MA (2013) Consistency of pharmaceutical products: an Karanakov L, Tonic-Ribarska J, Glavas-Dodov M, Trajkovic-Jolevska
FDA perspective on hot-melt extrusion process. Melt Extrusion. S (2012) Analysis and critical review of ICH Q8, Q9 and Q10
Springer, pp 435–445 from a generic pharmaceutical industry view point. J Geophys
Ha J-M, Seo J-W, Kim S-H, Kim J-Y et al (2017) Implementation of Res Solid Earth 93:10174–10190
quality by design for formulation of Rebamipide gastro-retentive Kaushal A, Arora S, Singh S, Sharma N (2020) Assessing the impact of
tablet. AAPS PharmSciTech 18(8):3129–3139 formulation variables on dissolution profile of sustained release
Ha E-S, Sim W-Y, Park S-M, Shin S et al (2018) Quality by Design tablet of metformin hydrochloride by quality by design approach.
(QbD): application of risk assessment. Korean Soc Food, Drug Res J Pharm Technol 13(5):2350–2358
Cosmet Regul Sci 13(2):85–97 Kelley B, Cromwell M, Jerkins J (2016) Integration of QbD risk
Hasnain MS, Ahmed SA, Khatoon A, Afzal M et al (2021) Pharmaceu- assessment tools and overall risk management. Biologicals
tical product development: a quality by design (QbD) approach. 44(5):341–351
In advances and challenges in pharmaceutical technology. Else- Kenett RS, Kenett DA (2008) Quality by design applications in biosim-
vier, pp 131–146 ilar pharmaceutical products. Accred Qual Assur 13(12):681–690
Jacques P, Hbid C, Destain J, Razafindralambo H et al (1999) Optimi- Kenjale PP, Joshi MA, Khatavkar UN, Dhapte VV et al (2020) Parox-
zation of biosurfactant lipopeptide production from Bacillus sub- etine hydrochloride push-pull osmotic pump tablets: designing
tilis S499 by Plackett-Burman design. In: Twentieth symposium an innovative, scalable push-pull osmotic drug delivery system
on biotechnology for fuels and chemicals. Springer using QbD approach. Drug Deliv Lett 10(2):104–116
Jagan B, Mahapatra AK, Murthy NP, Patra RK (2021), Quality by Khurana B, Arora D, Narang RK (2020) QbD based exploration of
Design (QbD): principles, underlying concepts and regulatory resveratrol loaded polymeric micelles based carbomer gel for
prospects. Thai J Pharm Sci (TJPS). 45(1) topical treatment of plaque psoriasis: In vitro, ex vivo and in vivo
Jain S (2014) Quality by design (QBD): a comprehensive understand- studies. J Drug Deliv Sci Technol 59:101901
ing of implementation and challenges in pharmaceuticals devel- Kim JY, Kwon K (2013) A study on the comparison of Korea GMP
opment. Int J Pharm Pharm Sci 6:29–35 with PIC/S GMP for enhancing international competecy of
Javed MN, Kohli K, Amin S (2018) Risk assessment integrated QbD medicinal product quality. Yakhak Hoeji 57(6):432–441
approach for development of optimized bicontinuous mucoad- Koeberle M, Schiemenz W (2017) QbD: improving pharmaceutical
hesive limicubes for oral delivery of rosuvastatin. AAPS Pharm- development and manufacturing workflows to deliver better
SciTech 19(3):1377–1391 patient outcomes. Pharm Technol 4(9):s20–s23
Jayagopal B, Shivashankar M (2017) Analytical quality by design–a Kolekar YM (2019) Understanding of DoE and its advantages in phar-
legitimate paradigm for pharmaceutical analytical method devel- maceutical development as per QbD approach. Asian J Pharm
opment and validation. Mech Mater Sci Eng J 9 Technol 9(4):271
Jayagopal B, Murugesh S (2020) QbD-mediated RP-UPLC method Kothari C, Patel R (2015) Regulatory need: harmonized PIC/S GMP,
development invoking an FMEA-based risk assessment to its overview and comparison with WHO GMP. Pharm Reg
Affairs 4(150):2

13
680 Journal of Pharmaceutical Investigation (2022) 52:649–682

Kovács B, Kovács-Deák B, Székely-Szentmiklósi I, Fülöp I et al (2021) Mukharya A, Chaudhary S, Mansuri N, Misra AK (2012a) Functional-
Quality-by-design in pharmaceutical development: From current ity advancement of poorly soluble biovariable anti hypertensive
perspectives to practical applications. Acta Pharm 71(4):497–526 drug by sophisticated sd-fbp technology as per enhanced qbd. Int
Krummen L (2015) Regulatory considerations for Implementation J Res Dev Pharm Life Sci 1(2):72–89
of the QbD Paradigm for biologics: laying the foundation for Mukharya A, Chaudhary S, Shah A, Mansuri N et al (2012b) Devel-
product and process lifecycle management. Quality by design opment and scale-up of SD-FBP formulation technology in line
for biopharmaceutical drug product development. Springer, pp with parametric QbD. Res J Pharm Sci 1(1)
693–705 Mundada PK, Sawant KK, Mundada VP (2017) Formulation and
Lawrence XY (2008) Pharmaceutical quality by design: product and optimization of controlled release powder for reconstitution for
process development, understanding, and control. Pharm Res metoprolol succinate multi unit particulate formulation using
25(4):781–791 risk based QbD approach. J Drug Deliv Sci Technol 41:462–474
Lawrence XY, Amidon G, Khan MA, Hoag MA et al (2014) Muntean AC, Negoi OI, Rus LL, Vonica AL et al (2020) Formulation
Understanding pharmaceutical quality by design. AAPS J of orodispersible tablets containing paracetamol and their in vitro
16(4):771–783 characterization–a QbD approach. FARMACIA 68(3):436–446
Lee J, Kim J (2018) QbD platform design based on edge computing for Murphy T, O'Mahony N, Panduru K, Riordan D et al (2016) Pharma-
bioequivalent drugs. In: Proceedings of the Korea Information ceutical manufacturing and the quality by design (QBD), process
Processing Society Conference. Korea Information Processing analytical technology (PAT) approach. In: 2016 27th Irish signals
Society and systems conference (ISSC). IEEE
Lee J, Kim J-E (2019) Application of open source based DoE R pro- Namjoshi S, Dabbaghi M, Roberts MS, Grice JE et al (2020) Quality
gram for the development of QbD. Yakhak Hoeji 63(5):274–281 by design: development of the Quality Target Product Profile
Lee S-H, Kim J-E (2021) Quality by Design Applied Development (QTPP) for semisolid topical products. Pharmaceutics 12(3):287
of Immediate-Release Rabeprazole Sodium Dry-Coated Tablet. Nayak B, Elchidana P, Sahu P (2017) A quality by design approach
Pharmaceutics 13(2):259 for coating process parameter optimization. Indian J Pharm Sci
Mandenius C-F (2009) Quality by design (QbD) for biotechnology- 79(3):345–352
related pharmaceuticals. Biotechnol J 4(6):600–609 Nayak AK, Ahmed SA, Beg S, Tabish M et al (2019) Application of
Mandlik SK, Agarwal PP, Dandgavhal HP (2020) Implementation of quality by design for the development of biopharmaceuticals. In:
Quality By Design (QBD) approach in formulation and develop- Pharmaceutical quality by design. Elsevier, pp 399–411
ment of ritonavir pellets using extrusion spheronization method. Negi P, Singh B, Sharma G, Beg S et al (2015) Biocompatible lidocaine
Int J Appl Pharm 12:139–146 and prilocaine loaded-nanoemulsion system for enhanced percu-
Markl D, Warman M, Dumarey M, Bergman E-L et al (2020) Review taneous absorption: QbD-based optimisation, dermatokinetics
of real-time release testing of pharmaceutical tablets: state-of-the and in vivo evaluation. J Microencapsul 32(5):419–431
art, challenges and future perspective. Int J Pharm 582:119353 Negi P, Singh B, Sharma G, Beg S et al (2016) Phospholipid micro-
Martinez-Marquez D, Terhaer K, Scheinemann P, Mirnajafizadeh A emulsion-based hydrogel for enhanced topical delivery of lido-
et al (2020) Quality by Design for industry translation: three- caine and prilocaine: QbD-based development and evaluation.
dimensional risk assessment failure mode, effects, and criticality Drug Deliv 23(3):941–957
analysis for additively manufactured patient-specific implants. Oh G-H, Park J-H, Shin H-W, Kim H-W et al (2018) Quality-by-design
Eng Rep 2(1):e12113 approach for the development of telmisartan potassium tablets.
Marto J, Gouveia L, Jorge I, Duarte A et al (2015) Starch-based Picker- Drug Dev Ind Pharm 44(5):837–848
ing emulsions for topical drug delivery: a QbD approach. Col- Ohage E, Iverson R, Krummen L, Taticek R et al (2016) QbD imple-
loids Surf B 135:183–192 mentation and post approval lifecycle management (PALM).
Marto J, Gouveia L, Gonçalves L, Gaspar L et al (2016) A quality Biologicals 44(5):332–340
by design (QbD) approach on starch-based nanocapsules: a Pallagi E, Ismail R, Paal T, Csóka I (2018) Initial risk assessment as
promising platform for topical drug delivery. Colloids Surf B part of the quality by design in peptide drug containing formula-
143:177–185 tion development. Eur J Pharm Sci 122:160–169
McDermott M, Chatterjee S, Hu X, Ash-Shakoor A et al (2015) Appli- Pallagi E, Jójárt-Laczkovich O, Németh Z, Szabó-Révész P et al (2019)
cation of Quality by Design (QbD) approach to ultrasonic atomi- Application of the QbD-based approach in the early development
zation spray coating of drug-eluting stents. AAPS PharmSciTech of liposomes for nasal administration. Int J Pharm 562:11–22
16(4):811–823 Patel H, Parmar S, Patel B (2013) A comprehensive review on Quality
Mesut B, Ȍzsoy Y, Aksu B (2015) The place of drug product critical by Design (QbD) in pharmaceuticals. Development 4:5
quality parameters in quality by design (QBD). Turk J Pharm Patel GM, Shelat PK, Lalwani AN (2017) QbD based development of
Sci 12(1):75–92 proliposome of lopinavir for improved oral bioavailability. Eur
Miller K (2020) Reconciling quality by design and transdermal product J Pharm Sci 108:50–61
development. Pharmaceutics 12:273 Patel D, Patel M, Soni T, Suhagia B (2021) Topical arginine solid
Mishra M, Raza K (2021) Design of experiments for the development lipid nanoparticles: development and characterization by QbD
of transdermal drug products. Design of experiments for phar- approach. J Drug Deliv Sci Technol 61:102329
maceutical product development. Springer, pp 57–67 Patil AS, Pethe AM (2013) Quality by Design (QbD): a new concept
Mishra SM, Rohera BD (2017) An integrated, quality by design (QbD) for development of quality pharmaceuticals. Int J Pharm Qual
approach for design, development and optimization of orally Assur 4(2):13–19
disintegrating tablet formulation of carbamazepine. Pharm Dev Patricio RP, Catai RE, Michaud RE, Nagalli A (2013) Model of risk
Technol 22(7):889–903 management based in the FMEA technique–a case study in the
Mistree RY, Chodhary UM, Shah CN, Upadhyay U. Quality by design construction of gabions. Electron J Geotech Eng 18:4183–4199
approach for pharmaceutical product development: a compre- Peltonen L (2018) Design space and QbD approach for production of
hensive review drug nanocrystals by wet media milling techniques. Pharmaceu-
Mohurle MSM, Asnani MDAJ, Chaple DR, Kurian MJ et al (2019) tics 10(3):104
Quality by Design (QbD): an emerging trend in improving qual-
ity & development of pharmaceuticals. SJMPS 05:1132–1138

13
Journal of Pharmaceutical Investigation (2022) 52:649–682 681

Pharm B, Pharm M (2013) Quality by design (QbD): manufactur- industrial biotechnology: bioprocess, bioseparation, and cell
ing and product quality of generics drugs perspective. J Global technology, pp 1–36
Trends Pharm Sci 4(4):1257–1262 Shah A, Jameel P, Patel SM (2015) Application of QbD principles
Pramod K, Tahir MA, Charoo NA, Ansari SH et al (2016) Pharma- for lyophilized formulation development. In: Quality by design
ceutical product development: a quality by design approach. Int for biopharmaceutical drug product development. Springer, pp
J Pharm Investig 6(3):129 137–158
Purohit P, Shah K (2013) Quality By Design (QBD): new parameter Shah B, Khunt D, Bhatt H, Misra M et al (2016) Intranasal delivery of
for quality improvement & pharmaceutical drug development. venlafaxine loaded nanostructured lipid carrier: risk assessment
Pharma Sci Monit 3(3) and QbD based optimization. J Drug Deliv Sci Technol 33:37–50
Puskeiler R, Kreuzmann J, Schuster C, Didzus K et al (2011) The Shah N, Mehta T, Gohel M (2017) Formulation and optimization of
way to a design space for an animal cell culture process accord- multiparticulate drug delivery system approach for high drug
ing to Quality by Design (QbD). In: BMC proceedings. BioMed loading. AAPS PharmSciTech 18(6):2157–2167
Central Sharma T, Jain A, Kaur R, Saini S et al (2020) Supersaturated
Raina H, Kaur S, Jindal AB (2017) Development of efavirenz loaded LFCS type III self-emulsifying delivery systems of sorafenib
solid lipid nanoparticles: risk assessment, quality-by-design tosylate with improved biopharmaceutical performance: QbD-
(QbD) based optimisation and physicochemical characterisation. enabled development and evaluation. Drug Deliv Transl Res
J Drug Deliv Sci Technol 39:180–191 10:839–861
Raman VVSSN, Useni Reddy M, Arunkanth Krishnakumar N, Sharma A, Jeyaprakash RM, Rinchi Bora AC (2021) Impact of qual-
Maheshwar Reddy M et al (2015) Quality improvement with ity risk management process in pharmaceutical industry to cur-
scientific approaches (QbD, aQbD and PAT) in generic drug tail the non-conformity
substance development: review. Int J Res Dev Pharm Life Sci. Sharmada A, Kakodkar SS (2015) Pharmaceutical Quality-by-Design
https://​doi.​org/​10.​4172/​2278-​0238.​10001​02 (QbD): basic principles
Rapalli VK, Banerjee S, Khan S, Jha PN et al (2021) QbD-driven Shekhawat P, Pokharkar V (2019) Risk assessment and QbD based
formulation development and evaluation of topical hydrogel optimization of an Eprosartan mesylate nanosuspension: in-
containing ketoconazole loaded cubosomes. Mater Sci Eng C vitro characterization, PAMPA and in-vivo assessment. Int J
119:111548 Pharm 567:118415
Rathore AS (2009) Roadmap for implementation of quality by Silva BM, Silva C (2021) Quality by design for nanocarriers. In:
design (QbD) for biotechnology products. Trends Biotechnol Nanoparticles for brain drug delivery. Jenny Stanford Publish-
27(9):546–553 ing, pp 351–382
Rathore AS (2014) QbD/PAT for bioprocessing: moving from theory Simões A, Veiga F, Vitorino C, Figueiras C (2018) A tutorial for
to implementation. Curr Opin Chem Eng 6:1–8 developing a topical cream formulation based on the quality by
Rathore AS (2016) Quality by design (QbD)-based process develop- design approach. J Pharm Sci 107(10):2653–2662
ment for purification of a biotherapeutic. Trends Biotechnol Singh L, Sharma V (2015) Quality by Design (QbD) approach in
34(5):358–370 pharmaceuticals: status, challenges and next steps. Drug Deliv
Rathore AS, Winkle H (2009) Quality by design for biopharmaceuti- Lett 5(1):2–8
cals. Nat Biotechnol 27(1):26–34 Singh SK, Venkateshwaran R, Simmons SP (2010) Oral controlled
Rathore AS, Mhatre R (2011) Quality by design for biopharmaceuti- drug delivery: Quality by design (QbD) approach to drug
cals: principles and case studies, vol 1. Wiley development. In: Oral controlled release formulation design
Rathore AS, Garcia-Aponte R, Golabgir A, Vallejo-Diaz BM et al and drug delivery, pp 279–303
(2017) Role of knowledge management in development and Singh R, Muzzio S, Ierapetritou M, Ramachandran R (2015) A
lifecycle management of biopharmaceuticals. Pharm Res combined feed-forward/feed-back control system for a QbD-
34(2):243–256 based continuous tablet manufacturing process. Processes
Raw AS, Lionberger R, Lawrence R (2011) Pharmaceutical equiva- 3(2):339–356
lence by design for generic drugs: modified-release products. Singh MK, Pharm M, Singh S. Faculty of Pharmacy, BBDNIIT Luc-
Pharm Res 28(7):1445–1453 know Quality-by-Design in Pharmaceutical Development
Rawal M, Singh A, Amiji MM (2019) Quality-by-design concepts to Sitre DG, Kamble RK (2021) Formulation development and evalu-
improve nanotechnology-based drug development. Pharm Res ation of new albendazole tablets with integrated Quality by
36(11):1–20 Design (QbD) approach. J Drug Deliv Ther 11(2):123–134
Saha M, Saha DR, Ulhosna DR, Sharker SM et al (2021) QbD based Sonam C, Rathore K. Quality by design. Risk 16(17):18
development of resveratrol-loaded mucoadhesive lecithin/chi- Soans D, Chandramouli R, Kavitha A, Roopesh S et al (2016) Appli-
tosan nanoparticles for prolonged ocular drug delivery. J Drug cation of design of experiments for optimizing critical quality
Deliv Sci Technol 63:102480 attributes (CQA) in routine pharmaceutical product develop-
Saitoh S (2018) The identification of Critical Quality Attributes ment. J Pharm Res 15(3):96–100
(CQAs) for the development of antibody drugs. Yakugaku Zasshi Sreedhara A, Wong RL, Lentz Y, Schoenhammer K et al (2015)
138(12):1475–1481 Application of QbD principles to late-stage formulation devel-
Santosh J, Audumbar SM, Gorakhnath H, Ashpak H. Quality by Design opment for biological liquid products. Quality by design for
(QbD) principles and applications in product development: an biopharmaceutical drug product development. Springer, pp
overview 115–135
Saydam M (2016) Association of QbD tools with biopharmaceutical Sri KR, Bhavani TS, Teja PSB et al (2021) An outline of quality
classification systems. Pharm Eng, p 79 by design in pharmaceutical development. J Pharm Sci Res
Schweitzer M, Pohl M, Hanna-Brown M, Nethercote P et al (2010) 13(4):200–205
Implications and opportunities of applying QbD principles to Stegemann S, Connolly P, Matthews W, Barnett R et al (2014) Applica-
analytical measurements. Pharm Technol 34(2):52–59 tion of QbD principles for the evaluation of empty hard capsules
Shah RB, Park M, Read EK, Khan MA et al (2009) Quality by design as an input parameter in formulation development and manufac-
(QbD), biopharmaceutical manufacture. In: Encyclopedia of turing. AAPS PharmSciTech 15(3):542–549

13
682 Journal of Pharmaceutical Investigation (2022) 52:649–682

Su Q, Bommireddy Y, Shah Y, Ganesh S et al (2019) Data recon- near infrared, and inline pellet size analysis. J Pharm Sci
ciliation in the Quality-by-Design (QbD) implementation of 109(12):3598–3607
pharmaceutical continuous tablet manufacturing. Int J Pharm Von Stosch M, Hamelink J-M, Oliveira R (2016) Hybrid modeling as a
563:259–272 QbD/PAT tool in process development: an industrial E. coli case
Suciu S, Iurian S, Bogdan C, Iovanov C et al (2018) QbD approach in study. Bioprocess Biosyst Eng 39(5):773–784
the development of oral lyophilisates with ibuprofen for paedi- Waghule T, Rapalli VK, Singhvi G, Manchanda P et al (2019) Vori-
atric use. Farmacia 66(3):514–518 conazole loaded nanostructured lipid carriers based topical deliv-
Suresh S, Roy S, Ahuja B (2015) Quality by design: an overview. ery system: QbD based designing, characterization, in-vitro and
Indian Drugs 52(02):5–11 ex-vivo evaluation. J Drug Deliv Sci Technol 52:303–315
Syed SM, More RI (2020) Quality by design: an approach for formula- Wang J, Kan S, Chen T, Liu J (2015) Application of quality by design
tion development. Inventi Rapid 1:1–6 (QbD) to formulation and processing of naproxen pellets by
Sylvester B, Porfire A, Achim M, Rus L et al (2018) A step forward extrusion–spheronization. Pharm Dev Technol 20(2):246–256
towards the development of stable freeze-dried liposomes: Wen H, Park K (2011) Oral controlled release formulation design and
a quality by design approach (QbD). Drug Dev Ind Pharm drug delivery: theory to practice. Wiley, Hoboken
44(3):385–397 Xu X, Khan MA, Burgess DJ (2011) A quality by design (QbD)
Thombre NA, Ahire PS, Kshirsagar SJ (2021) Formulation develop- case study on liposomes containing hydrophilic API: I. for-
ment and evaluation of mouth dissolving tablet of aspirin by mulation, processing design and risk assessment. Int J Pharm
using QbD approach. Dhaka Univ J Pharm Sci 20(1):19–29 419(1–2):52–59
Thoorens G, Krier F, Leclercq B, Carlin B et al (2014) Microcrystal- Yamashita H, Sun CC (2020) Material-sparing and expedited devel-
line cellulose, a direct compression binder in a quality by design opment of a tablet formulation of carbamazepine glutaric acid
environment—a review. Int J Pharm 473(1–2):64–72 cocrystal–a QbD approach. Pharm Res 37(8):1–10
Uhlenbrock L, Sixt M, Strube J (2017) Quality-by-Design (QbD) Yu X, Lawrence XY, Teng Y, Gaglani DK et al (2019) Implementation
process evaluation for phytopharmaceuticals on the example of of pharmaceutical quality by design in wet granulation. In: Hand-
10-deacetylbaccatin III from yew. Resource-Efficient Technol book of pharmaceutical wet granulation. Elsevier, pp 703–733
3(2):137–143 Zandieh M (2020) GMP risk assessment at an organic and natural food
Vanaja K, Shobha Rani R (2007) Design of experiments: concept and production plant with emphasis on infrastructure by PHA and
applications of Plackett Burman design. Clin Res Regul Aff FMEA methods. World 9(1):11–15
24(1):1–23 Zhang L, Mao S (2017) Application of quality by design in the current
Verma U, Naik JB, Patil JB, Yadava SK (2017) Screening of pro- drug development. Asian J Pharm Sci 12(1):1–8
cess variables to enhance the solubility of famotidine with Zurdo J, Arnell A, Obrezanova O, Smith N et al (2015) Early imple-
2-HydroxyPropyl–β-Cyclodextrin & PVP K-30 by using Plack- mentation of QbD in biopharmaceutical. BioMed Res Int. https://​
ett-Burman design approach. Mater Sci Eng C 77:282–292 doi.​org/​10.​1155/​2015/​605427
Vivekanandan S (2019) Quality by design in pharmaceutical formula-
tion. In: Computer applications in drug discovery and develop- Publisher's Note Springer Nature remains neutral with regard to
ment. IGI Global. pp 221–239 jurisdictional claims in published maps and institutional affiliations.
Vo AQ, Kutz SK, He H, Narala S et al (2020) Continuous manufac-
turing of ketoprofen delayed release pellets using melt extru-
sion technology: application of QbD design space, inline

13

You might also like