You are on page 1of 32

Drug Development and Industrial Pharmacy

ISSN: 0363-9045 (Print) 1520-5762 (Online) Journal homepage: http://www.tandfonline.com/loi/iddi20

Quality-by-design approach for development of


telmisartan potassium tablets

Ga-hui Oh, Jin-Hyun Park, Hye-Won Shin, Joo-Eun Kim & Young-Joon Park

To cite this article: Ga-hui Oh, Jin-Hyun Park, Hye-Won Shin, Joo-Eun Kim & Young-Joon
Park (2017): Quality-by-design approach for development of telmisartan potassium tablets, Drug
Development and Industrial Pharmacy, DOI: 10.1080/03639045.2017.1414233

To link to this article: https://doi.org/10.1080/03639045.2017.1414233

Accepted author version posted online: 18


Dec 2017.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=iddi20

Download by: [Gothenburg University Library] Date: 19 December 2017, At: 03:54
Quality-by-design approach for development of
telmisartan potassium tablets
Ga-hui Oh, Jin-Hyun Park, Hye-Won Shin, Joo-Eun Kim†, and Young-Joon Park††

College of Pharmacy, Ajou University, Suwon, Korea


Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

††

t
Corresponding author: College of Pharmacy, Ajou University, Suwon City 443-749, Korea

ip
Phone: +82-31-219-3493, Fax: +82-31-219-3432, E-mail: parkyj64@gmail.com

cr

us
Corresponding author: Yuhan R&D institute, Yongin City, Korea

Phone: +82-31-899-4052, Fax: +82-31-275-6145, E-mail: 77jooeun@naver.com


an
M
ed
pt
ce
Ac
Abstract

A quality-by-design approach was adopted to develop telmisartan potassium (TP) tablets, which

were bioequivalent with the commercially available Micardis® (telmisartan free base) tablets. The

dissolution pattern and impurity profile of TP tablets differed from those of Micardis® tablets

because telmisartan free base is poorly soluble in water. After identifying the quality target product

profile and critical quality attributes (CQAs), drug dissolution and impurities were predicted to be

risky CQAs. To determine the exact range and cause of risks, we used the risk assessment (RA) tools,

PHA and FMEA to determine the parameters affecting drug dissolution, impurities, and formulation.
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
The range of the design space was optimized using the face-centered central composite design

among the design of experiment (DOE) methods. The binder, disintegrant, and kneading time in the

cr
wet granulation were identified as X values affecting Y values (disintegration, hardness, friability,

us
dissolution, and impurities). After determining the design space with the desired Y values, the TP
an
tablets were formulated and their dissolution pattern was compared with that of the reference tablet.

The selected TP tablet formulated using design space showed a similar dissolution to that of
M

Micardis® tablets at pH 7.5. The QbD approach TP tablet was bioequivalent to Micardis® tablets in
ed

beagle dogs.

Keywords: Telmisartan potassium, Quality-by-Design, Design of Experiment, Risk Assessment,


pt

Design Space, Bioequivalence.


ce
Ac
Abbreviations

QbD : Quality by Design

TP : Telmisartan Potassium

QTPP : Quality Target Product Profiles

CQA : Critical Quality Attributes

RA : Risk Assessment
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

CMA : Critical Material Attributes

t
ip
CPP : Critical Process Parameters

cr
PHA : Preliminary Hazard Analysis

FMEA : Failure Mode and Effect Analysis us


an
DoE : Design of Experiments
M

CCD: Central Composite Design


ed

FCCCD: Face-centered Central Composite Design


pt

API : Active Pharmaceutical Ingredients


ce

RPN : Risk Priority Numbers (RPNs = P × S × D).


Ac

P : Probability, D : Detectability, and S : Severity.

USP : United States Pharmacopeia


1. Introduction

Telmisartan, 4-[(2-n-propyl-4-methyl-6-(1-methylbenzimidazole-2-yl)-benzimidazole-

1yl)methyl]-biphenyl-2-carboxylic acid, is a selective angiotensin II type 1 receptor (AT1R) blocker,

which reduces blood pressure [1]. It was developed by Boehringer Ingelheim and launched in 1999

as Micardis®. Telmisartan free base has been mainly used in commercial products. Owing to its low

solubility in water (BCS class II), its oral bioavailability is poor [2]. The solubility of telmisartan

free base has been increased by the addition of sodium salt using sodium hydroxide (NaOH) and

meglumine as alkylating agents [3]. In contrast, telmisartan potassium (TP, a salt form) is highly
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

soluble, and thus, it exhibits a different dissolution pattern.

t
ip
Most drugs are either weak organic acids or bases and, therefore, can exist in different salt forms.

cr
Although the active pharmaceutical ingredient (API) is the same in all these salts, they could be

us
considered as distinct chemical entities with their own chemical and biological profiles [4]. To

improve the general properties of APIs such as solubility and stability, their novel salts are often
an
introduced. Numerous pharmaceutical technologies for enhancing the solubility of insoluble drugs
M

use formulations such as amorphous forms (solid dispersions/cocrystals), polymeric micelles,

inclusion complexation, size reduction including nanonization [5], self-microemulsifying drug


ed

delivery systems (SMEDDS), surfactant/co-solvent systems [6], solid lipid nanoparticles

(SLN)/liposomes, pH-modified form, and salt forms [7]. However, one of the strategies for
pt

improving solubility and stability is the incorporation of different salt forms of an approved API into
ce

the marketed brand dosage form of a company, followed by bioequivalence assessment [4].
Ac

To develop a bioequivalent formulation with the salt alternative of telmisartan free base, we used a

quality-by-design (QbD) approach [8], which involves building quality into products by design [9].

The QbD [10] approach ensures that the pharmaceutical development process is focused on

ultimately providing a scientific understanding of how process parameters affect product

performance. This knowledge enables the establishment of a design space, product, and process

specifications, as well as appropriate manufacturing controls that ensure the consistent achievement

of all necessary quality targets and product requirements [11]. Hence, QbD emphasizes the
systematic development of pharmaceutical products based on sound scientific principles and refers

to the achievement of predictable quality with desired, predetermined specifications [12].

The process and formulation can be understood by developing them based on the multivariate

analysis of designed experiments, historical data, or both that identify and characterize the critical-

to-quality process parameters as well as the root causes of variability [13]. Design of experiments

(DOE) is a useful tool for identifying and optimizing critical process parameters (CPPs)/critical

material attributes (CMAs) [14]. To optimize the process and formulation, statistical DOEs [15] are

used during the pharmaceutical product/process development [16]. In particular, response surface

methodologies, including statistical experimental designs such as central composite design and Box-
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
Behnken design, have been commonly used [17, 18, 19].

ip
The aim of this study was to develop a novel alternative TP salt with bioequivalence to telmisartan

cr
free base using a QbD approach. After defining the quality target product profile (QTPP) and critical

us
quality attributes (CQAs), drug dissolution and impurities in the process were predicted to be the

risky CQAs. To determine the exact range and cause of the risk, we used the risk assessment (RA)
an
tool preliminary hazard analysis (PHA) [20] and failure mode effect analysis (FMEA) [21] to
M

identify the parameters that affect drug dissolution, impurities, and formulation [22]. The range of

design space was optimized using the face-centered central composite design, which is one of the
ed

DoE methods [23]. Furthermore, the drug dissolution and bioavailability of the TP tablet were
pt

evaluated in beagle dogs and compared with those of Micardis® tablet(telmisartan free base).
ce

The tablets of this novel TP salt were significantly smaller than the marketed tablets, thereby

increasing patients’ compliance, especially in elderly patients with hypertension who face difficulties
Ac

in swallowing.
2. Materials and methods

2.1 Materials

TP was provided by Hwail Pharmaceutical Co., Ltd. (Seoul, Korea). Magnesium oxide (MgO) was

purchased from Tomita Pharmaceutical Co., Ltd (Tokushima, Japan). Potassium chloride was

purchased from Samchun Chemical Co., Ltd. (Pyeongtaek-si, Korea). D-mannitol was purchased

from Merck & Co., Inc. (Kenilworth, NJ, USA). Microcrystalline cellulose and croscarmellose

sodium were purchased from JRS Pharma Co., Ltd. (Patterson, NY, USA). Crospovidone was

purchased from BASF Pharma Co., Ltd., (Rotherstraße, Berlin, Germany). Colloidal silicon dioxide
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
was purchased from Evonic Health Care Co., Ltd., (Rellinghauser Straße, Essen, Germany).

cr
Magnesium stearate was provided by Merck KGaA (Frankfurter StraBe, Darmstadt, Germany). All

other chemicals were of reagent grade and obtained commercially.

us
an
2.2 Defining QTPP

To determine the QTPP of the TP tablet, we specified the indication; dosage form, design, and
M

strength; the route of administration, pharmacokinetics, shelf life, and primary packaging.
ed

Furthermore, these items were also justified in detail from the perspectives of pharmacists,

physicians, and patients. In addition, the research goals and risk of justification according to
pt

pharmacists, physicians, and patients were specified (Table 1).


ce

2.3 Determining CQAs


Ac

To identify the CQAs of the TP tablet, the quality attributes such as physical characteristics,

appearance, identification, dissolution, impurities, weight variation, content uniformity, assay, and

residual solvents were specified. Furthermore, the acceptable values of the various items and the

CQAs were specified. The results of the CQA estimation are shown in green, yellow, and red colors

depending on the risk (Table 2). Thus, the CQA colors in the Table 2 were justified and evaluated

according to safety, efficacy, and quality specifications as the three major parameters of medicines.
The CQA targets are similar to the criteria specified in the certificate of analysis (CoA); however,

in contrast, the CQA list includes items to determine whether the QA is general or critical. Among

the various QAs, CQAs were identified by color, and the QA feasibility was described in terms of

safety, efficacy, and quality.

The “Justification” section in the CQA table is written as; content and content uniformity test may

affect safety and efficacy; currently, these process variables and excipients may affect the content

and content uniformity of the TP product. Therefore, content and content uniformity tests should be

performed using formulation studies and process development; thus, they were selected as CQAs.

However, the “Identification test” in the CQA table did not affect the formulation studies and
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
process variables of the TP tablets because only the presence of the API was confirmed. Therefore, it

is not a CQA but only a general quality attribute.

cr
2.4 RA of CMAs and CPPs
us
an
An RA was performed to select and evaluate the CMAs and CPPs that affect the CQAs. In this

study, PHA and FMEA were used as RA tools [24]. PHA consist of a table in which each item is
M

indicated by a color: green, yellow, or red (low-, medium-, or high-risk, respectively). Green

indicates a broadly acceptable risk and no further investigation is needed while yellow indicates an
ed

acceptable risk level, which may require further investigation/justification to reduce the probability.
pt

Finally, red indicates an unacceptable risk that requires further investigation to reduce the risk.
ce

Among the various MAs, the functional groups of excipients and APIs that are considered to affect

CQAs are listed in the PHA and FMEA Tables. For the FMEA analysis, each failure mode was
Ac

ranked using an estimated frequency of probability (P), the probability of failure undetected later in

the detectability (D), and severity (S). The P of detection and S are scored using a scale of 1–4. P is

defined as the frequency of a specific failure that can result in failure mode. It should refer to the P

of the cause, which is a specific failure mode for a particular effect/event.

The score based on the P of occurrence of failure is divided into unlikely (1), occasional (2),

repeated (3), and regular (4). The scores based on the likely S effect on a drug product with CQAs
were divided into minor (1), moderate (2), major (3), and extreme (4).The score based on the P of

failure of detection is divided into always detected (1), regularly detected (2), likely not detected (3),

and normally not detected (4). Failure risks were calculated using risk priority numbers (RPNs),

which are the product of P, S, and D (RPNs = P × S × D). The values of P, S, and D for each failure

mode in the case of the TP tablets were assigned based on previous experience and literature reviews,

and the RPNs were calculated.

A high RPN value for the concentration of disintegrant indicated that it could introduce a lag time

in dissolution. The potential cause could be a low disintegrant concentration with a high occurrence

value (P = 4), a low detectability value (D = 2), and an extreme severity value (S = 4); the calculated
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
RPN for this failure mode was 32. Failure modes with RPNs ≥ 30 were set as criteria for

ip
considering the DoE as a possible failure.

cr
2.5 Preparation of TP tablets
us
an
The TP tablets were prepared according to the formulation compositions shown in the range of

design space or preliminary studies using the wet granulation method with a high shear mixer. The
M

TP tablets each comprised 91.1 mg TP, 30.0 mg magnesium oxide (MgO), 10.0 mg potassium

chloride, 151.9 mg microcrystalline cellulose (MCC), 20.0 mg D-mannitol, 20.0 mg croscarmellose


ed

sodium, 10.0 mg crospovidone, 10.0 mg colloidal silicon dioxide, and 7.0 mg magnesium stearate.
pt

TP, the API, and part of the colloidal silicon dioxide were mixed for 5 min at 15 rpm using a
ce

laboratory mixer. The obtained mixture, MgO, D-mannitol, crospovidone, and part of the MCC were

further mixed for 8 min at 15 rpm using a laboratory mixer, and then the obtained mixture was
Ac

filtered through a 20-mesh screen. For the wet granulation process, an ethanol solution was added to

this mixture as a binding solution to yield wet granules, and the impeller and chopper speed (150 and

2,000 rpm, respectively) were maintained during the wet granulation process using a high shear

mixer for 5 min. Then, the granules were dried using a fluid bed dryer (Glatt GPCG1, Ramsey, NJ,

USA) at 55°C for 1.5 h until the water content was < 2.0%. The dried granules were passed through

a 0.065-inch screen processed using a Cone Mill instrument (Hosokawa micron Ltd., Cheshire, UK).
The milled granules were mixed with croscarmellose sodium, potassium chloride, part of the MCC,

and part of the colloidal silicon dioxide for 3 min and then finally blended with magnesium stearate.

TP tablets were prepared by compression using a rotary tablet machine to a targeted hardness and

mass weight of 11 kp and 350 mg, respectively, as indicated by the design space.

2.6 Evaluation of TP tablets

2.6.1 Hardness test

The hardness level of the TP tablets was assayed using the Model 8M bench top hardness tester

(Pharmatron, Aesch, Switzerland). The tablet was placed between the edges of the fixed and
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
movable part of the instrument, and the hardness was measured in kiloponds.

ip
cr
2.6.2 Friability

us
The friability of the TP tablets was assayed using a FT 2 friability tester (Pharmatron model FT

2 friability tester, Aesch, Switzerland). Friability is the measure of tablet strength, and it was
an
calculated as a percentage using the following formula:
M

Friability (%) = [(w1 - w2)/w1] × 100,

where w1 and w2 are the weights of the tablets before and after the test.
ed
pt

2.6.3 Disintegration test


ce

The disintegration rate of the TP tablets was assayed using a tablet disintegration tester

(Pharmatron model DisiTest 20 tablet disintegration tester, Aesch, Switzerland). The disintegration
Ac

test was performed according to the US Pharmacopeia (USP) by placing six TP tablets into a tablet

disintegration tester.

2.6.4 In vitro dissolution of TP tablets

The dissolution test was performed using the USP XXXII dissolution apparatus II with 900 mL

of 20 mM potassium dihydrogen phosphate buffer at pH 7.5 ± 0.1 as the dissolution medium at 37 ±


0.5°C. The speed of the paddle was adjusted to 75 rpm. The TP and Micardis® tablets at an

equivalent dose of 80 mg telmisartan base were placed into a dissolution tester (Agilent dissolution

tester, Santa Clara, CA, USA). After 30 min or at predetermined intervals, 4 mL of the medium was

sampled and filtered through a membrane filter (0.45-μm). The concentration of telmisartan in the

filtrate was analyzed using ultraviolet (UV) spectrophotometer (Agilent 8453 UV/visible [Vis]

spectrophotometer, Santa Clara, CA, USA) at 296 nm. Then, the in vitro dissolution profiles of the

TP tablets and reference drug (Micardis®) were compared using the similarity factor (f2)

recommended for dissolution profile comparison in the Guidance for Industry of US Food and Drug

Administration (FDA).
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
2.6.5 High-performance liquid chromatography (HPLC) analysis

cr
The content and impurity level of telmisartan were assayed using a high-performance liquid

us
chromatography (HPLC) system equipped with a separation module and a UV/Vis detector (Agilent
an
1200 series, Santa Clara, CA, USA). To assay telmisartan content, a Luna C18 column, (150 × 3.9

mm, 5 μm; Phenomenex Inc., Torrance, CA, USA) was used as the stationary phase. The mobile
M

phase, composed of 20 mM potassium dihydrogen phosphate:methanol (4:6, v/v, pH 3.0 ± 0.05),

was filtered through a 0.22-μm membrane filter and degassed using an online degasser. The column
ed

temperature, detection wavelength, eluent flow rate, and injection volume were 40°C, 237 nm, 1.5
pt

mL/min, and 20 µL, respectively. Calibration samples were prepared by accurately weighing and
ce

dissolving 32.0 mg telmisartan to obtain a stock solution, which was subsequently diluted to

concentrations of 32.0–320 μg/mL. The calibration curve was rectilinear with a correlation
Ac

coefficient of 0.999, whereas the standard deviation (SD) of the accuracy and precision was < 2%.

To assay telmisartan impurity levels, a Zorbax SB C18 column (50 × 4.6 mm, 3.5 μm; Agilent

Inc.) was used as the stationary phase. Mobile phases A and B were composed of 18 mM ammonium

dihydrogen phosphate (pH 3.0 ± 0.1):methanol in the ratios of 4:6 and 2:8 (v/v), respectively. The

following gradient program was followed: 0% B (0–12 min), 0–20% B (12–13 min), 20–90% B (13–

15 min), 90% B (15–25 min), and 90–0% B (25–30 min). The column temperature, detection
wavelength, eluent flow rate, and injection volume were 35°C, 298 nm, 1.2 mL/min, and 10 µL,

respectively.

2.7 Development of the design space

The Minitab® software (version 18; Minitab Inc., State College PA, USA) was used for

performing the DoE. Three input (X) values were selected [two types of CMAs (binding solution

and a disintegrant) and one type of CPP (granulation time) through the PHA/FMEA of RA]. The

levels were tested at a narrow range in the knowledge space based on the preliminary studies.

The DoE was set in the range of 40 to 100 mL binding solution, 2 to 6% disintegrant agent, and 3
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
to 7 min of wet granulation (kneading) time. Thus, the output (Y) value was selected as the most

ip
important variable to optimize the disintegrant, hardness, friability, drug dissolution, and impurities

cr
as a factor affected by the three X values.

us
A face-centered center composite design(FCCCD) was used for the DoE to optimize the CQAs

according to the selected CPPs and CMAs. The experimental plan of 20 experimental runs (23 + [2 x
an
3] + 6) was obtained and performed randomly. The values of the predetermined dependent variables
M

were obtained and statistically analyzed. The main, interaction, and quadratic effects of the factors

related to the properties of the TP tablets were evaluated and the effect of each independent variable
ed

on the dependent variables was investigated. Variables with p-values < 0.05 were considered
pt

statistically significant. Each factor was tested at three different levels, and six center points were

included. The Minitab® software was used for the design, analysis, and plotting of the various three-
ce

dimensional (3D) and contour plots. With the models developed and validated, the design space was
Ac

finally set through all individual acceptance regions for each CMA and CPP.

2.8 Bioequivalence in beagle dogs

Ten Canis familiaris (beagle dogs) were randomly divided into two groups, and the reference

drug (Micardis®) and TP tablets were orally administered under fasting conditions using a two-

period single-dose crossover design. There was a 1-week drug washout period between each dosing.
After oral drug administration, blood samples (3.0 mL) were collected from the jugular vein at 0,

0.25, 0.5, 1, 2, 4, 6, 8, 12, 24, 36, 48, and 72 h. The blood samples were immediately centrifuged at

3,000 rpm for 10 min, and the plasma was stored at -70°C until the analysis. The plasma

concentration of telmisartan in beagle dogs administering the reference drug (Micardis®) or the TP

tablets was analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). The

Waters® Micromass® Quattro Premier XE Tandem quadrupole mass spectrometer (Quattro Premier

XE, Waters, Milford, MA, USA), interfaced with an ultraperformance LC (UPLC, Alliance HT

2795, Waters, Milford, MA, USA), and equipped with a Unison UK-C18 column (75 mm × 2.0 mm

internal diameter, 3 μm, Imtakt, Ringoes, NJ, USA) was used.


Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
The stock solution of telmisartan was prepared by adjust the concentration of 200 μg/mL in

ip
methanol, which was subsequently diluted with the blank serum to a concentration range of 10, 30,

cr
50, 100, 500, 1000, 5,000, and 20,000 ng/mL as a plasma standard. The standard solutions and test

us
solutions were prepared by adding 10 μL of the internal standard solution (50 μg/mL of telmisartan-

d7 in 50% methanol) to 50 uL of each standard plasma and test plasma. After adding 1000 μL of
an
acetonitrile, centrifuging for 10 minutes at 3,000 rpm for 1 minute with vortexing, 2 μL of
M

supernatant was added to LC-MS/MS. A 2 μL sample was separated chromatographically using

isocratic conditions for the mobile phase (A:B, 35:65, v/v). The mobile phase A was 0.1% formic
ed

acid in 10 mM ammonium formate in distilled water and mobile phase B was 99.99% acetonitrile.
pt

The flow rate was set at 0.35 mL/min. The calibration curve was drawn with the telmisartan peak
ce

area ratio to the peak area ratio of the internal standard (of telmisartan-d7) obtained. We have

performed assay validation with specificity, linearity, accuracy and precision. The calibration curve
Ac

was rectilinear with a correlation coefficient of 0.99849 to 0.99937 while the SD of the accuracy and

precision was < 2%.

Statistical analysis

Pharmacokinetic parameters, including total area under the plasma concentration-time curve from

time zero to infinity (AUC0–∞), half-life (t1/2), maximum plasma drug concentration (Cmax), and time
to reach Cmax (Tmax) were calculated using the WinNonlin® software (Pharsight Co., Cary, NC, USA)

of Crystal Genomics Pharmaceutical Co., Ltd.


Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
cr
us
an
M
ed
pt
ce
Ac
3. Results and Discussion
3.1 Defining QTPP

QTPP is a summary of the quality characteristics or attributes of a drug product that need to be

achieved to ensure its safety and efficacy [25]. It forms the basis for product development design.

As shown in Table 1, the indication; dosage form, design, and strength; the route of administration,

pharmacokinetics, shelf life, and primary packaging were specified. All items except the route of

administration, indication, and residual solvents showed risky justification as QTTP elements

between the TP and reference tablets. This indicates that the requirements for pharmaceutical

equivalence with Micardis® as a reference drug were an approximately similar or the same dosage
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
form, design, and strength; the route of administration; pharmacokinetics; shelf life; and primary

ip
packaging according to pharmacists, physicians, and patients' perspectives. However, the QTPP may

cr
be updated or revised at various stages of development as new information is obtained during the

process [25].
us
an
3.2 Determining CQAs
M

As shown in Table 2, the quality attributes of the TP tablet such as physical attributes, appearance,

identification, dissolution, impurities, weight variation, content uniformity, assay, and residual
ed

solvents were specified. This indicated that the requirements for pharmaceutical equivalence
pt

between Micardis® as a reference drug and the TP tablets were approximately or the same quality of
ce

attributes: physical characteristics, appearance, identification, dissolution, impurities, weight

variation, content uniformity, assay, and residual solvents with respect to safety, efficacy, and
Ac

quality. However, dissolution and impurities were deemed the CQAs of the TP tablet.

Failure to meet the dissolution specification would affect the bioavailability and efficacy while

both formulation and process variables affect the dissolution profile. This CQA was investigated

throughout the formulation and process development in the quality RA and design space.

Degradation products can affect safety and must be controlled based on compendia/International

Council for Harmonisation ICH requirements or reference product characterization to limit patient
exposure[8]. Formulation and process variables may affect degradation products. Therefore, the

degradation products were evaluated during product and process development in the quality RA and

design space[23].

3.3 Quality RA of CMAs and CPPs affecting CQAs

The PHA and FMEA were performed based on previous experience and preliminary

experiments[22]. CMA has a critical effect on the CQA among several MAs. CPP refers to the

critical effect of CQAs among several PPs. The PHA and FMEA of the MAs were included in the

analysis and RPNs were calculated for each parameter (Table 3). The PHA and FMEA of PPs were
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
included in the analysis and RPNs were calculated for each parameter (Table 4).

ip
In the group of core-related factors, wet granulation agents (ethanol as a binding solution) and

cr
superdisintegrants (croscarmellose sodium and crospovidone) were estimated. Table 3 shows the

us
selection of CMAs from several MAs using PHA and FMEA. The risk factors that must be

addressed in the DoE selected by the RPN calculation were identified as “binding solution” and
an
“disintegrant.” Table 4 also shows the results of the screening of CPPs among several PPs using
M

PHA and FMEA. The risk factor that must be addressed in the DoE selected by the RPN calculation

was confirmed as the “time of the granulation” (the kneading time).


ed
pt

3.4 Development of design space

The binding solution, disintegrant agent, and granule kneading time, which are the critical factors
ce

influencing CQAs were selected as CMAs and CPPs at the risk assessment stage. The experimental
Ac

runs with independent variables and corresponding responses for the 20 formulations tested are

presented in Table 5. We used a three-factor, three-level face-centered composite design, which

consisted of replicated center points and a set of points lying at the midpoints of each edge of a

multidimensional cube that defined the area of interest. The center composite design method is a

reactive surface design method that can estimate the first and second terms effectively and model the

curvature of the response variables by adding the experimental points to the previous factor
design[19]. The face-centered composite design is the central composite design which, unlike a

general centered composite design, has all the axis points on but not outside the face in a rectangular

parallelepiped. As shown in Table 5, the number of total experiments conducted was 23 + (2 × 3) + 6

= 20 because it occurred at the face and vertex of the rectangular parallelepiped.

For optimized modeling of the DoE, the response surface design analysis using the regression

equation and the ANOVA was optimized by selecting only the significant factors. Finally, we

determined whether the model was adequate using a lack of fit test. The P-value was > 0.05 and, so,

we could not reject the null hypothesis for lack of conformity. Therefore, the DoE modeling was
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

optimized, and then the results could be interpreted using a program such as a Pareto chart and

t
ip
residual plots for each of the five reaction values (Y 1–5 values) in Figure 1.

cr
As shown in the factorial plots of Figure 2, the results could be interpreted based on whether or not

us
the binding solution, disintegrant agent, and granulation kneading time affected the disintegration

(Y1), hardness (Y2), friability (Y3), drug dissolution at 30 min (Y4), and impurities after 3 months
an
(Y5). In addition, the graph of the 2D contour plots and the 3D response surface plots (Figure 2)
M

demonstrated how the binder, disintegration, and granulation time affected each reaction Y value.

In the contour plot and main effect plot of hardness (Y2), we observed that the hardness increased
ed

with increasing amounts of binding solution, but the amount of disintegrant agent had little effect on
pt

hardness. However, the contour plot and main effect plot of disintegration (Y1) showed that the

disintegration rate depended more on the specific disintegrant concentration (rate) than it did on the
ce

amount of binding solution. The contour plot and main effect plot of drug dissolution (Y4) showed
Ac

that the specific disintegrant concentration (rate), the amount of binding solution, and kneading time

affected the dissolution rate. In addition, contour plots for impurities (Y5) showed that degradation

products depended on the amount of disintegrant and binding solution, rather than on the time of

granulation.

Figure 3 shows the optimum design value of the actual operating space by superimposing the

contour plots shown in Figure 2. By overlaying the contour maps from each response on top of each
other, RSM was used to identify the ideal window of operability design space per proven acceptable

range and edges of failure with regard to individual goals.

For the design space, the range for contour plots of disintegrant was set up such that the tablet

disintegration was not more than 10 min. The range of the contour plots of hardness was set at 10.0–

11.5 kp. The range of the contour plots of friability was set up such that the minimum friability was

not more than 0.15%. The range of the contour plots of disintegrant was set such that the maximum

dissolution after 30 min was not less than 90%. The range of the contour plots of impurities was set

such that the minimum impurities after 3 months was not less than 0.15%.

The knowledge space area, which was derived from the preliminary studies of the disintegrant
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
agent (X2), was in the range of 2% to 6%. However, the range of disintegrant that satisfied the target

ip
values of the five individual reaction values was 3.0–4.5%, but it is different from the actual usage

cr
range. Therefore, it is possible to set the control space area to 3.0–4.0% because the values need to

remain at this level in terms of dissolution and impurities.


us
The knowledge space area, which was derived from the preliminary studies of the binding solution
an
(X1), was in the range of 40–100 mL. However, the range of binding solution that satisfied the target
M

values of the five individual reaction values was 57.0–75.0 mL, but it was different from the actual

range. Therefore, the control space area should be between 60.0 and 70.0 mL considering the
ed

dissolution and impurities.


pt

The knowledge space area, which was derived from the preliminary studies of the kneading time

(X3), was in the range of 3.0–7.0 min. However, the range of kneading time that satisfied the target
ce

values of the five individual reaction values was in the range of 3.5 to 6.0 min, but it differed from
Ac

the actual usage range. Therefore, the control space area must be between 4.0 and 5.5 min,

considering the dissolution and impurities.

3.5 In vitro dissolution of TP tablets

After preparing the final formulation in the range of the design space, the drug dissolution test was

performed on the TP and the commercial Micardis® reference tablets. Figure 4 shows the drug
release profiles of the TP and Micardis® reference tablets were similar. The dissolution rate of the

TP and Micardis® reference tablets in the pH 7.5 medium was 28.4 ± 4.26 and 26.7 ± 2.39, 61.0 ±

5.31 and 56.1 ± 2.50, 82.9 ± 4.96 and 76.9 ± 2.5, 100.1 ± 0.49 and 97.5 ±0.57, and 100.7 ± 0.42 and

98.1 ± 0.40 at 5, 10, 15, 30, 45 min, respectively. Furthermore, the similarity of the results of the in

vitro dissolution tests was confirmed by the f2 value [4], with 50 < f2 < 100 indicating the similar

dissolution pattern of the two products. The f2 of the TP and Micardis® reference tablets was 69.6 in

the pH 7.5 medium in the standard and test methods based on the USP monograph. Thus, the TP
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

tablets and Micardis® reference drugs showed a similar correlation of dissolution profiles in the pH

t
ip
7.5 medium of the standard and test methods in the USP monograph. Furthermore, the results of the

cr
in vitro dissolution tests in the pH 7.5 medium of the standard and test methods indicated that both

us
the TP tablets and Micardis® reference drugs completely dissolved within 30 min by up to 70%.
an
Therefore, the TP tablets had a comparable dissolution with the Micardis® reference drug.
M

3.7 Bioequivalence in beagles


ed

The in vivo pharmacokinetic properties of TP after oral administration in beagle dogs were
pt

evaluated. The TP concentrations assessed from the plasma concentration-time profiles of Micardis®
ce

and the TP tablets at a dose of 8.0 mg/kg are presented in Table 6 and Figure 5. After oral
Ac

administration of various drug formulations to beagles, the AUC of the TP and Micardis® tablets

(20,838.2 ± 7,044.3 and 21,285.1 ± 8,340.3 ng·h−1·mL−1, respectively) were not significantly

different (P > 0.05), which indicates that they had similar absorption and circulation times in the

blood. The 90% confidence interval (CI, 0.9071–1.0814) of the geometric least square mean ratios of
the AUC was 0.9790 of the point estimate, which was within 0.80–1.25 of the bioequivalence

criteria for telmisartan.

The Cmax values of the TP and Micardis® tablets (7,925.2 ± 1,662.0 and 8,315.5 ± 1,675.9

ng/mL, respectively) were not significantly different (P > 0.05), which indicates that they had similar

absorption rates in the blood. Furthermore, the 90% CI (0.8598–1.0976) of the Cmax was 0.9531 of

the point estimate, which was within the 0.80–1.25 range of the bioequivalence criteria of

telmisartan. These results indicate that the TP and Micardis® tablets have similar absorption rates
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
following oral administration.

ip
Table 6 shows that the 90% CI of the geometric mean ratios (test/reference drug) of the C max was not

cr
within the acceptable range in the in vivo experiment in beagles. Therefore, the preclinical study

us
demonstrated that the reference and test product were bioequivalent, indicating that the test drug
an
could be a suitable alternative for the commercial product (Micardis®).
M

4. Conclusion
ed

We developed TP tablet that was bioequivalent to the commercial Micardis® tablet using a QbD
pt

approach. Telmisartan free base, which is indicated for the treatment of hypertension has a different
ce

dissolution pattern from that of TP because it is poorly soluble in water.


Ac

After listing the QTPP and CQA, rapid drug dissolution was predicted as a risky CQA. To

determine the exact range and cause of risk according to the CMAs and CPPs, we used PHA and

FMEA to determine the parameters that affect drug dissolution and formulation. The range of the

design space was optimized using the face-centered central composite design among the available

DoE methods. The binding solution, disintegrant agent, and granule manufacturing time in the wet
granulation methods were identified as X values affecting Y values (disintegration time, hardness,

friability, drug dissolution at 30 min, and impurities after 3 months). The design space of the desired

hardness, degree of grinding, elution, impurities, and disintegration time was determined, the tablets

were prepared, and then the dissolution patterns of the TP and reference tablets were compared.

The selected TP tablets comprised telmisartan potassium, magnesium oxide, potassium chloride,

microcrystalline cellulose, D-mannitol, croscarmellose sodium, crospovidone, colloidal silicon

dioxide, and magnesium stearate. The TP tablets were manufactured using wet granulation because
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
of the dissolution properties. The TP tablet selected through the design space showed a similar

ip
dissolution rate to that of the Micardis® tablet at pH 1.2. Finally, the TP tablets were bioequivalent to

cr
the Micardis® tablets in beagles. Thus, the TP tablet appears to be a promising candidate

formulation. us
an
Moreover, the TP tablet of novel alternative salt was significantly smaller than the Micardis® tablet
M

of telmisartan free base. Therefore, it may improve patients’ compliance, especially in elderly
ed

patients with hypertension, who have trouble in swallowing.


pt

Acknowledgement
ce

This work was supported by the Technological Innovation R&D Program (C0400223) funded by the
Ac

Small and Medium Business Administration (SMBA, Korea).


REFERENCES

1. Pitt B, Konstam MA. Overview of angiotensin II-receptor antagonists. The American journal
of cardiology. 1998;82(10):S47-S49.
2. Sekar V, Chellan VR. Immediate release tablets of telmisartan using superdisintegrant-
formulation, evaluation and stability studies. Chemical and Pharmaceutical Bulletin.
2008;56(4):575-577.
3. Tran PHL, Tran HTT, Lee B-J. Modulation of microenvironmental pH and crystallinity of
ionizable telmisartan using alkalizers in solid dispersions for controlled release. Journal of
Controlled Release. 2008;129(1):59-65.
4. Cho KH, Choi H-G. Development of novel bepotastine salicylate salt bioequivalent to the
commercial bepotastine besilate in beagle dogs. Drug development and industrial pharmacy.
2013;39(6):901-908.
5. Kim J-e, Park Y-J. Paclitaxel-loaded hyaluronan solid nanoemulsions for enhanced treatment
efficacy in ovarian cancer. International journal of nanomedicine. 2017;12:645.
6. Kim J-E, Park Y-J. High paclitaxel-loaded and tumor cell-targeting hyaluronan-coated
nanoemulsions. Colloids and Surfaces B: Biointerfaces. 2017;150:362-372.
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

7. Kalepu S, Nekkanti V. Insoluble drug delivery strategies: review of recent advances and

t
ip
business prospects. Acta Pharmaceutica Sinica B. 2015;5(5):442-453.
8. Guideline IHT. Pharmaceutical development. Q8 (2R) As revised in August. 2009.
9. Visser JC, Dohmen WM, Hinrichs WL, et al. Quality by design approach for optimizing the

cr
formulation and physical properties of extemporaneously prepared orodispersible films.
International journal of pharmaceutics. 2015;485(1):70-76.
10.

11.
services. Simon and Schuster; 1992.
us
Juran JM. Juran on quality by design: the new steps for planning quality into goods and

Rathore AS, Winkle H. Quality by design for biopharmaceuticals. Nature biotechnology.


an
2009;27(1):26-34.
12. McKenzie P, Kiang S, Tom J, et al. Can pharmaceutical process development become high
tech? AIChE Journal. 2006;52(12):3990-3994.
M

13. Rahman Z, Zidan AS, Habib MJ, et al. Understanding the quality of protein loaded PLGA
nanoparticles variability by Plackett–Burman design. International journal of pharmaceutics.
2010;389(1):186-194.
14. Salazar J, Heinzerling O, Müller RH, et al. Process optimization of a novel production
ed

method for nanosuspensions using design of experiments (DoE). International journal of


pharmaceutics. 2011;420(2):395-403.
15. Wu H, White M, Khan MA. Quality-by-Design (QbD): An integrated process analytical
pt

technology (PAT) approach for a dynamic pharmaceutical co-precipitation process


characterization and process design space development. International journal of
ce

pharmaceutics. 2011;405(1):63-78.
16. Lawrence XY. Pharmaceutical quality by design: product and process development,
understanding, and control. Pharmaceutical research. 2008;25(4):781-791.
Ac

17. Zidan AS, Sammour OA, Hammad MA, et al. Quality by design: Understanding the
formulation variables of a cyclosporine A self-nanoemulsified drug delivery systems by
Box–Behnken design and desirability function. International journal of pharmaceutics.
2007;332(1):55-63.
18. Lee Y-L, Kim M-S, Park M-Y, et al. Quality by design: understanding the formulation
variables and optimization of metformin hydrochloride 750 mg sustained release tablet by
Box–Behnken design. Journal of Pharmaceutical Investigation. 2012;42(4):213-220.
19. Park S-J, Choo G-H, Hwang S-J, et al. Quality by design: screening of critical variables and
formulation optimization of Eudragit E nanoparticles containing dutasteride. Archives of
pharmacal research. 2013;36(5):593-601.
20. Rausand M. Preliminary hazard analysis. Norwegian University of Science and Technology.
2005.
21. Stamatis DH. Failure mode and effect analysis: FMEA from theory to execution. ASQ
Quality Press; 2003.
22. Guideline IHT. Quality risk management. Q9, Current step. 2005;4:408.
23. Hakemeyer C, McKnight N, John RS, et al. Process characterization and design space
definition. Biologicals. 2016;44(5):306-318.
24. Kaljević O, Djuriš J, Djurić Z, et al. Application of failure mode and effects analysis in
quality by design approach for formulation of carvedilol compression coated tablets. Journal
of Drug Delivery Science and Technology. 2016;32:56-63.
25. Rathore AS. Roadmap for implementation of quality by design (QbD) for biotechnology
products. Trends in biotechnology. 2009;27(9):546-553.
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
cr
us
an
M
ed
pt
ce
Ac
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

Ac
ce
pt
ed
M
an
us
cr
ip
t
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

Ac
ce
pt
ed
M
an
us
cr
ip
t
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

Ac
ce
pt
ed
M
an
us
cr
ip
t
Table 1. Quality target product profile (QTPP)
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
cr
us
an
M

TP, telmisartan potassium; USP, United States Pharmacopoeia; RSD, relative standard deviation; CI,
ed

confidence interval; PK, pharmacokinetics; AUC0–t, area under the plasma concentration-time curve
pt

from time 0 to t; AUC0–, area under the plasma concentration-time curve from time 0 extrapolated to
ce

infinity; Cmax, maximum plasma concentration; API, active pharmaceutical ingredient; NMT, not
Ac

more than; NLT, not less than; green color, low risk; yellow color, medium risk; red color, high risk
Table 2. Critical quality attributes (CQAs)
Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
cr
us
an
QA, quality attribute; TP, telmisartan potassium; USP, United States Pharmacopoeia; RSD, relative
M

standard deviation; NMT, not more than; NLT, not less than; green color, low risk; yellow color,

medium risk; red color, high risk


ed
pt
ce
Ac
Table 3. (A) Qualitative preliminary hazard analysis (PHA) and (B) quantitative failure mode

effects analysis (FMEA) of material attributes (MAs)


Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
cr
us
an
M

TP, telmisartan potassium; CQA, critical quality attribute; CMA, critical material attribute; QTPP,
ed

quality target product profile; PSD, particle size distribution; RPN, risk priority number.
pt

Each failure mode in FMEA was ranked for estimated probability/frequency of occurrence (P), the
ce

probability that the failure would remain undetected later in the process (D), and severity (S) on a
Ac

scale of 1–4
Table 4. (A) Qualitative preliminary hazard analysis (PHA) and (B) quantitative failure mode

effects analysis (FMEA) of processing parameters (PPs)


Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
cr
us
an
M
ed

CQA, critical quality attribute; CPP, critical process parameter; QTPP, quality target product

profile; RPN, risk priority number.


pt

Each failure mode in FMEA was ranked for estimated probability/frequency of occurrence (P), the
ce

probability that the failure would remain undetected later in the process (D), and severity (S) on a
Ac

scale of 1–4
Table 5. Critical material attributes (CMA) and critical process parameters (CPP) affecting

critical quality attributes (CQAs)

CMA CPP CQA

X1 Y4 Y5
X2 Y1
Binding X3 Y2 Drug Impurities
Disintegrant Disintegrati Y3
Run solution Granulation Hardness dissolution (%) after
concentratio on time Friability (%)
volume time (min) (kp) after 30 min three months
n (%) (min)
(mL) (%)

1 40 2 3 10 7.20 0.22 95 ± 1.2 0.029

2 100 2 3 15 12.60 0.08 73 ± 3.3 0.065

3 40 6 3 6 7.05 0.23 98 ± 2.1 0.151

4 100 6 3 9 11.55 0.09 81 ± 2.2 0.320

5 40 2 7 12 8.40 0.17 80 ± 0.4 0.038


Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
6 100 2 7 18 14.55 0.05 69 ± 5.2 0.089

ip
7 40 6 7 7 8.85 0.18 87 ± 0.8 0.153

8 100 6 7 10 14.10 0.05 77 ± 2.2 0.339

cr
9 70 4 5 5 7.95 0.22 97 ± 1.4 0.080

us
10 70 4 5 12 13.05 0.06 79 ± 2.7 0.190

11 70 2 5 15 12.15 0.09 88 ± 3.5 0.046

12 70 6 5 9 10.20 0.13 97 ± 2.4 0.238


an
13 70 4 3 7 9.60 0.15 86 ± 4.3 0.135

14 70 4 7 10 12.45 0.09 94 ± 4.2 0.143


M

15 70 4 5 8 11.70 0.11 94 ± 2.2 0.139

16 70 4 5 7 11.40 0.11 96 ± 1.1 0.138


ed

17 70 4 5 8 10.65 0.12 96 ± 1.2 0.135

18 70 4 5 8 11.40 0.11 95 ± 1.7 0.135

19 70 4 5 9 10.80 0.12 93 ± 1.4 0.136


pt

20 70 4 5 9 11.25 0.10 97 ± 1.2 0.139


ce
Ac
Table 6. Pharmacokinetic parameters of telmisartan after oral administration as Micardis®

and TP tablets (80 mg) in beagle dogs

Micardis® tablet TP tablet1)


Pharmacokinetic Point
90% CI
parameters estimate
Mean ± SD*

Tmax (h)** 0.75 ± 0.21 0.85 ± 0.3 - -

Cmax (ng/mL) 8,315.5 ± 1,675.9 7,925.2 ± 1,662.0 0.9531 0.8598–1.0976

AUC(last) (ng∙h-1∙mL-1) 21,285.1 ± 8,340.3 20,838.2 ± 7,044.3 0.9790 0.9071–1.0814

AUC(inf) (ng∙h-1∙mL-1) 21,644.2 ± 8,861.4 21,024.2 ± 7,210.6 0.9713 0.9204–1.1302


Downloaded by [Gothenburg University Library] at 03:54 19 December 2017

t
ip
T1/2 (h) 13.9 ± 7.4 11.4 ± 5.7 - -
Data are presented as mean ± SD (n = 12).

cr
1
p < 0.05 compared with TP tablet group; CI, confidence interval; Tmax, time to reach

us
maximum plasma concentration; Cmax, maximum plasma concentration; AUClast, area

under the plasma concentration-time curve at the last time point; AUC0–, AUC
an
extrapolated to infinity; T1/2, half-life
M
ed
pt
ce
Ac

You might also like