Chavan A. V, Gandhimathi R. Quality by Design Approach: Progress in Pharmaceutical Method Development and Validation. Biomed Pharmacol J 2023;16(3).
Manuscript received on :22-09-2022
Manuscript accepted on :02-02-2023
Published online on: 21-07-2023
Plagiarism Check: Yes
Reviewed by: Dr. Vijay Kumar
Second Review by: Dr. Hind Shakir
Final Approval by: Dr. H Fai Poon

How to Cite    |   Publication History
Views Views: (Visited 354 times, 1 visits today)   Downloads PDF Downloads: 153

Avinash V. Chavan1 and R. Gandhimathi1*

Department of Pharmaceutical Chemistry and Analysis, School of Pharmaceutical Sciences, Vels Institute of Science, Technology and Advanced Studies(VISTAS), Pallavaram, Chennai-600117, Tamil Nadu, India.

Corresponding Author E-mail: avinashchavan9696@gmail.com

DOI : https://dx.doi.org/10.13005/bpj/2745

Abstract

Pharmaceutical analysis plays a significant role in pharmaceutical formulation quality assurance and control. Due to the pharmaceutical industries' rapid expansion and the production of pharmaceuticals all over the world, there is a greater need for novel analytical procedures in this sector. Establishing the identification, purity, physical properties, and potency of medications as well as the medication's bioavailability and stability is the goal of analytical method development. A few new drug applications were recently given regulatory flexibility by the Food and Drug Administration for an analytical method based on quality by design. With Quality by design, product design and development are performed methodically. Analytical methodologies have similar opportunities for implementing Quality by design as production procedures do. It consequently enhances formulation design, development efficiency, and capacity. The underpinnings of the QbD approach have been explored in this article due to their use in the creation and validation of analytical procedures. Additionally, a summary of experimental studies reporting the application of the QbD methodology to method development is included.

Keywords

Design of Expert; Method Development; Pharmaceutical Analysis; Quality by Design; Validation

Download this article as: 
Copy the following to cite this article:

Chavan A. V, Gandhimathi R. Quality by Design Approach: Progress in Pharmaceutical Method Development and Validation. Biomed Pharmacol J 2023;16(3).

Copy the following to cite this URL:

Chavan A. V, Gandhimathi R. Quality by Design Approach: Progress in Pharmaceutical Method Development and Validation. Biomed Pharmacol J 2023;16(3). Available from: https://bit.ly/46VMAvE

Introduction

Pharmaceutical industrial production is one of the most carefully regulated and governed sectors by traditional regulatory agencies, as the quality of pharmaceuticals is directly tied to public health. Consequently, it is necessary to control the quality of medications. The pharmaceutical industry aims to provide products and production processes that reliably meet established requirements. The ability to meet the demands and expectations of the client in terms of service, product, and process is what is meant by quality [1]. All regulatory organizations for pharmaceutical products place a high value on quality. Customer happiness equates to quality [2].

To prove that new medications are safe and effective, the pharmaceutical industries put lot of efforts into developing, producing, and bringing them to market [3]. They also work hard to comply with regulatory regulations. Every year, more medications are released onto the market. These medications could either be entirely new or structurally modified versions of already existing ones. From the moment a drug is released onto the market to the time, it is included in Pharmacopoeias; there is frequently a lag in time. This is brought on by potential risks associated with long-term and widespread use of these medications, reports of novel toxicities (leading to their removal from the market), the emergence of patient resistance, and rival companies’ launch of superior medicines. Standards and analytical techniques for certain medications may not be included in the pharmacopoeias under these circumstances. Thus, the need to create newer analytical methods for such medications arises [4]. A new strategy for medication development might boost productivity, offer regulatory clearance and flexibility, and bring about significant economic gains throughout the product’s life cycle [3].

Quality assurance and quality control of pharmaceutical formulations and bulk pharmaceuticals rely heavily on pharmaceutical analysis. The demand for novel analytical techniques in the pharmaceutical industries has increased due to the pharmaceutical industries’ rapid expansion and the manufacture of drugs in different parts of the world. The biopharmaceutical and vaccine industries use analytical techniques for research and development as well as to manage the inputs and outputs of manufacturing [5]. Establishing the identification, purity, physical properties, and potency of pharmaceuticals, as well as the drug’s bioavailability and stability, is the goal of analytical technique development. The improvement of analytical tools has led to recent developments in analytical methodologies. The development of better analytical methods and tools has resulted in shorter analysis times, greater precision and accuracy, and lower analysis costs. As a result, the majority of pharmaceutical companies are spending enormous sums of money to create cutting-edge analytical laboratories [4].

Product development heavily relies on the development and validation of analytical methods. In addition to guaranteeing that a drug’s quality is reached as per its intended therapeutic use, each stage of the product development life cycle includes a purity check that is performed using a trustworthy analytical approach. The analytical method utilized for the production of commercial products must be quick, dependable, and accurate since the ultimate quality check results of the finished piece and other batch data influence when the product can be released to the market. Analytical techniques frequently include estimating the target substance’s physical, chemical, physicochemical, and/or biological properties. Because they have many advantages over other non-chromatographic methods, chromatographic analytical techniques like High-performance liquid chromatography (HPLC), Gas chromatography (GC), and High-performance thin-layer chromatography (HPTLC), and supercritical fluid chromatography (SFC) are widely used. They need fewer samples and are sturdy and adaptable. These methods reduce the likelihood of human error by using automation.

The development of an analytical procedure that precisely serves the intended function is the analytical chemist’s top priority. There are currently two methods used for developing analytical methods in analytical chemistry. The former relies on trial and error and analyses one factor at a time (OFAT), in which a single parameter is optimized for the anticipated response while all other parameters are held constant. This procedure consistently results in the method’s narrow robust behaviour for the instrumental variables used throughout the method development phase. Because of this, developing analytical methods with the OFAT approach involves a significant chance of method failure and constantly necessitates the development of alternate methods or revalidation protocols, which drives up the cost of the technique [1].

Quality by design approach

Dr. Joseph M. Juran, a quality pioneer, is credited with creating the idea of quality by design (QbD). According to Dr. Juran, quality should be built into a product from the start, and most quality crises and issues originate from poor product design [6]. The QBD is described as “A systematic approach to development that begins with established objectives and stresses product and process understanding and process control, based on strong science and quality risk management”[7] in accordance with the International Conference on Harmonization Quality guideline 8 (ICH Q8) criteria. To improve robust production processes, facilitate product quality, and create products in accordance with “six sigma,” the concept of “quality by design” (QbD) has been developed in the pharmaceutical business [8].

According to several experts, the prospects for applying QbD to analytical methods are comparable to those for production processes [9]. The AQbD (analytical QBD) assists in the development of a trustworthy and reasonably priced analytical method that is relevant across the lifecycle of the product in order to enhance the regulatory leeway in the analytical approach. It refers to the ability to modify a method’s parameters anywhere in its design space, also known as the method operable design region (MODR) [10,11].

Objectives of Quality by design

Pharmaceutical QbD is a methodical approach to development that emphasizes both process and product comprehension and control based on solid science and quality risk management [6,12]. The following objectives of pharmaceutical QbD may be present:

Establishing clinical performance-based meaningful product quality specifications

To increase process capability and minimize product variability and faults by improving product / process design, understanding, and control.

To boost productivity in product development and production

To improve post approval change management and root cause analysis

Robustly developed products and processes are necessary for achieving this goal. The identification and management of issues affecting the quality of the drug product can also be facilitated by increased product and process expertise. The procedure should be improved after receiving regulatory approval to decrease product variability, flaws, rejections, and recalls.

Product design and development are done systematically with QbD. As a result, it improves formulation design, development speed, and capabilities. Additionally, it moves resources from an upstream proactive mode to a downstream corrective way. It enhances the manufacturer’s capacity to pinpoint the underlying reasons behind manufacturing failures. Therefore, improving manufacturing and product development efficiency is pharmaceutical QbD’s third goal.

Key Elements of Quality by design

In a pharmaceutical QbD strategy for product innovation, a claimant identifies qualities that are crucial to quality, changes them into the critical quality attributes (CQAs) of the drug product, and identifies the association between formulation/manufacturing factors and CQAs to ensure the patient will receive a medication product with these CQAs. The elements that make up QbD are as follows:

A quality target product profile (QTPP) that lists the drug product’s crucial quality attributes (CQAs) [6,13].

To support drug labelling and drug development efforts, TPP describes the necessary profile or attributes of a drug product. TPP lists the intended application, target, pace of administration, and other key product characteristics, together with quality designing for a drug product [14].

The phrase “TQPP” for product quality may be a logical extension of “TPP.” The QTPP is a crucial document that enables the rationalization and evolution of the data that is not inheritable throughout the drug’s lifespan. To affirm the targeted quality, a prospective outline of the attributes of quality for a drug product that will be reached while taking into account the safety and effectiveness of the targeted product is provided. Indefinite-quantity, type, strength, instrumentation closure system, identity, indefinite-quantity type, purity, and stability are all included in TQPP [15].

The QTPP is a possible list of the characteristics of a drug product that should be met in order to ensure that it is of the desired quality, in addition to the safety and efficacy of the medicinal product. The QTPP serves as the design framework for creating the product [6,12].

To ensure the required product quality, a CQA should be within appropriate limits. Clinical safety and efficacy, manufacturing attribute, and parameter boundaries approach edge of failure are examples of quality attributes [16].

A drug product’s quality may or may not be essential. The severity of the patient’s harm determines how critical of an attribute it is. The criticality of an attribute is unaffected by the probability of occurrence, detectability, or controllability [6,12].

Product design and understanding, including the identification of critical material attributes (CMAs)

Clinical research confirms that the product’s design impacts whether it can satisfy patients’ needs. Stability studies, which corroborate this, show that product design also affects whether a product can retain its performance throughout its shelf life. This kind of product insight might have avoided some historical stability breakdowns.

Creating a high-quality product that can deliver the necessary QTPP over the duration of its shelf life is the primary goal of product design and understanding. Designing a product can take many different directions because it is so open-ended. The following are crucial components of product design and comprehension:

The drug substance’s physical, chemical, and biological characterization (s))

Determining and selecting the excipient kind and grade, as well as being aware of the inherent excipient variability

Connections between drugs and excipients

Formulation optimization and CMA identification for both excipients and the medicinal substance

CMAs differ from CQAs in that they are used for input materials such excipients and drug ingredients. CQAs, on the other hand, apply to output materials like completed drug products and product intermediates [17].

3. Process design and comprehension include identifying critical process parameters (CPPs) and having a solid grasp of scale-up principles that connect CMAs and CPPs to CQAs.

When all significant causes of variation are recognized and explained, variability is controlled by the process, and product quality attributes can be predicted with reasonable accuracy, a process is often regarded as being well understood. The input operating parameters like mixing time, stirring speed, etc., of unit operation are called process parameters. A process parameter should be monitored or managed to guarantee that the process yields the desired quality when variability affects a crucial quality feature.

The formation of a control plan with three tiers of controls, as follows, is the result of the knowledge gathered from properly structured development studies:

The CQAs of the output materials are continuously monitored at Level 1 using automatic engineering control. The most adaptable level of control is this one. Pharmaceutical control at level 2 includes adjustable material attributes, process parameters within the defined design area, and fewer end-product tests. The typical level of control employed in the pharmaceutical sector is Level 3.This control method is based on rigorous end-product testing, closely controlled material properties, and process-related parameters. Any significant change in these necessitates regulatory control due to the incomplete categorization of the causes of variability and the lack of knowledge regarding CMAs and CPPs’ effect on the CQAs for medicinal products. The formulation of acceptance criteria, the necessity for further controls, and the debate about acceptable variability consume a significant amount of industry and regulatory resources. A hybrid strategy that combines levels 1 and 2 can be applied. A control strategy is described by ICH Q8 (R2) as a planned set of controls that are drawn from current product and process knowledge and ensure process efficiency and product quality.

4. Process capability and ongoing development

Process capacity measures the intrinsic variability of a stable process under statistical control with regard to the established acceptance criteria. Through continuous improvement programmes that focus on removing sources of significant variance from the process operation conditions and raw material quality, process capability can be used to measure process improvement. When significant deviations are detected, remedial and preventive actions must be implemented; this can be done by regularly checking process data for Cpk and other statistical process control metrics.

Regulatory Perspective of QbD

Regulatory bodies now strongly emphasize QbD alone rather than only “Quality by Testing” or “Quality by Chance”. Analytical procedures are a crucial component of the control plan concerning the pharmaceutical quality system (ICH Q 10 recommendations). Analytical QbD will be implemented in the manufacturing process to guarantee predetermined performance and product quality as a control technique [7].

ICH guidelines and QBD

The ICH guidelines provide clear definitions of QbD principles: Q8 (R1): pharmaceutical development, Q9: quality risk management, and Q10: pharmaceutical quality system.

Stages in QBD vs AQBD

Analytical QbD implementation follows a similar method to that of product QbD. Initially, the target measurement for implementing AQbD is dependent on the product file in the form of the ATP (analytical target profile) and CQA (ATP is the analogue of QTPP in product design).A comparison between QBD and AQBD is given in Table 1.

Table I: Parallels between QbD for Process and Product, and AQbD.

QbD

AQbD

Involves quality target product profile (QTPP)

Involves analytical target profile (ATP)

CQAs related to patients requirement or product development

CQAs related to analytical method development

Consider design space

Consider method operable design region

(MODR)

Consist of process

performance qualification (PPQ)

Method validation

Implementation AQBD

Analytical Target Profile (ATP)

 ATP specifies the objective of the development of analytical methods. The definition of ATP, recently offered by PhRMA and EFPIA, is as follows: “ATP is a declaration that describes the method’s goal and is used to guide method selection, design, and development activities.” Following regulatory authorities’ approval of the ATP statement, ATP is a crucial AQbD characteristic that enables increased improvement of analytical techniques and their selection. While the examples above are mostly focused on directly measurably and changeable technique parameters, the ATP should ideally cover all important aspects of method performance. [20,21].

Analytical Method Performance Characteristics

These are specified to satisfy the requirements of the analytical target profile. For chromatographic separations, USP and ICH have published numerous validation factors and are regarded as method performance characteristics. Accuracy, specificity, linearity, precision, detection limit, and quantification limit are these parameters. Robustness and range.

Selection of Analytical Techniques

The chosen analytical methodology must meet the validation requirements of ICH [8] as well as the required method performance specified in Adenosine triphosphate (ATP).

Risk Assessment

The parameters that affect the ATP are identified by risk assessment as the essential method variables. Following the identification of the technique, AQbD concentrates on developing the method and includes a thorough evaluation of the risks related to variability, such as analytical techniques, instrument settings, measurement and methodology parameters, sample properties, sample preparation, and ambient factors. The ICH Q9 guideline must be followed in the risk assessment strategy: Risks to the quality over the product lifecycle are assessed, controlled, communicated, and reviewed using a systematic approach [22].

Design of Experiments

Method operable design region (MODR) can be formed in the method development phase, which could serve as a source for reliable and affordable methods, in compliance with the requirement of ICH Q8 recommendations, regarding “design space” in product development. DoE implementation during the method development phase necessitates a deep comprehension of input variable selection and output reaction. The following are the components of  DoE in the AQbD technique.

Screening

Screening allows for the exclusion of qualitative input characteristics. It lists the different critical method parameters (CMP) that should be considered during the optimization studies. The CMP that has to be regulated or subjected to DOE approaches in MODR optimization should be separated as a result of the screening studies.

Optimization

Quantitative metrics for critical methods in variables (i.e., CMP) can be introduced at this point either directly from risk assessment or through screening. It provides a basis for comprehending the scientific connection between the quantities of input variables (CMP) and responses at the output, which will significantly impact the approach’s effectiveness and ATP.

Selection of DOE Tools

Numerous methods can be employed throughout the optimization to derive a statistical correlation (model). The quantity of input variables, acquaintance with regulated parameters, and scientific knowledge of the relationship between outcome and variable (if any) must all be taken into account while selecting the tool for DoE.

Surface Response Plots

Counter (2D) or Surface response plot (3D) represents the impact of input variables on output variables. Numbers like −1, 0, and +1, in both axes, represent the coded level of variables used in DOE.

Model Validation

Before choosing from a contour or graph, the results of an actual experimental run must verify the expected values for the desired technique response. The model must then undergo regression analysis in order to be statistically validated.

Application of AQBD for method development and validation

Numerous papers using the DOE methodology for developing analytical methods have already been published. For the testing of various bulk pharmaceuticals or active pharmaceutical ingredients, methods for HPLC, UPLC analysis, etc., are developed with high accuracy and precision. These QBD-steered approaches have also been used to determine the number of pharmaceuticals in various dosage forms, including tablets, capsules, and vesicular drug delivery systems, such as liposomes, cubosomes, exosomes, ethosomes, etc. Most research teams today use pharmacological models to validate their in vivo results. As a result, the validity of in vitro results is uncertain in the absence of in vivo research. In these situations, estimating the drug concentration in plasma samples or any other bodily fluids is desirable. This problem requires a suitable analytical technique with a higher sensitivity for detecting the minute-to-minute concentrations in fluids. The AQBD methodology primarily produces the analytical method’s robust performance.

Testing the stability profile of pharmacological compounds is an intriguing need of the analytical approach. The safety and effectiveness of the therapeutic product are impacted by the chemical stability of pharmaceutical molecules, which is a significant problem. A drug product may encounter several situations during storage times that could cause the product to degrade over time. In these situations, it is preferred to use analytical techniques to detect the degradation products. Understanding a molecule’s stability facilitates the choice of an appropriate formulation and packaging and the provision of proper storage conditions and shelf life, all of which are necessary for regulatory paperwork. The market withdrawal of medications is prevented by an accurate stability indication assay or identification of the degradation products of drugs or formulations.

Before submitting a registration dossier, stability tests of novel drug moieties are now required. Long-term (12 months) and accelerated stability investigations are also included in the stability studies (6 months). However, intermediate studies (6 months) can be carried out under more hospitable circumstances than those employed in rapid studies. Therefore, it would take considerably longer to analyze degradation products using separation, identification, and quantification methods. Forced degradation studies help produce degradants in a much shorter time than stability experiments, often a few weeks. To establish a stability-indicating approach that can later be used for examining samples produced by accelerated and long-term stability tests, forced degradation samples can be used [22].Some of the exciting Research works involving the use of the QBD approach for HPLC method development are summarized in Table 2.


Drug

Analytical technique

Mobile phase

Experimental design

Independent variables

Dependent variables

Reference

Abiraterone acetate

RP-HPLC Method

CAN/phosphate buffer (20:80 %v/v)

Box-Behnken

Mobile phase composition, pH, and flow rate

Retention time and peak area

[23]

Amiodarone hydrochloride

HPLC

ACN/MeOH/buffer ( 4.6/3.4/2)

QBD approach

 

Mobile phase pH, % organic phase, and column temperature

[24]

Zolmitriptan, naratriptan, dihydroergotamine, ketotifen, and pizotifen

RP-HPLC

24
Factorial design

ACN% in the mobile phase, mobile-phase pH, nature of the buffer, and column temperature

Resolution and Run time

[25]

Artesunate and

Amodiaquine impurities

Green HPLC method

Ethanol and 10mM acetic acid

pH, temperature, and gradient slope

3-level full factorial design

[26]

Atorvastatin

RP-HPLC

Acetonitrile: water (50: 50)

Box–Behnken statistical design

Mobile phase (acetonitrile: water), flow rate (Rt), and UV wavelength

 Area of the chromatogram (AUC), retention time (Rt, min), and tailingfactor (%)

[27]

Ceftazidime

RP-HPLC

ACN to acetic acid (75:25)

Face-centred cubic design

Mobile phase ratio(ACN) and flow rate

Peak area (PA), retention time (Rt), theoretical plate count (TPC),and tailing factor (TF)

[28]

Ceftriaxone

Sodium

RP-HPLC

Acetonitrile to water (0.01% triethylamine with pH 6.5) (70:30, v/v),

Central composite design

Mobile phase composition and pH

Retention time, theoretical plate, and peak asymmetry

[29]

Daclatasvir

HPLC, LC-MS/MS, UPLC

55% buffer and 45% ACN

Central composite design

pH and temperature

Resolution of impurity (c-h) and drug

[30]

Efavirenz

HPLC

Methanol, 10 mM ammonium acetate buffer (70:30 v/v),

32 full factorial design

Flow rate  and pH of the buffer

Retention time (y1) and peak area

[31]

Eltrombopag olamineand its degradation products

Stability-indicatingRP-HPLC/ RP-UPLC

0.1 % trifluoroacetic acid (TFA) and acetonitrile

24 factorial design

Column temperature, flow rate, the organic ratio in mobile phase, and the concentration of TFA

Resolution

[32]

Etofenamate

RP-HPLC

Methanol and 0.2% triethylamine in water at 85:15

Central composite design

pH of aqueous

phase, percentage of

the aqueous phase, and

flow rate

Retention time

[33]

Ferulic acid

RP-HPLC

ACN: water (47:53 % v/v

27 Taguchi design, face-centred composite design

Mobile phase ratio (X1) and flow rate

Peak area (PA), retention time (RT), tailing factor

[34]

Ketoprofen

Stability-indicating

RP-HPLC

Phosphate buffer–methanol (50: 50v/v)

Central composite design

Mobile phase ratio and pH of mobile phase

Theoretical plates and peak tailing

[35]

Nevirapine

Reversed-phase HPLC bioanalytical method

68:9:23% v/v elution of methanol, acetonitrile, and water

Box–Behnken design

Mobile phase ratio, pH, and flow rate

Peak area, retention time, theoretical plates, and peak tailing

[36]

Olmesartan medoxomil

Stability-Indicating HPLC

Acetonitrile and water

(40 : 60 v/v)

Face-centred cubic design

Mobile phase ratio and flow rate

Peak area, retention time, theoretical plates and peak tailing

[37]

Rufinamide

RP – HPLC bioanalytical method

Buffer :ACN at 84.7:15.3% v/v

Box Behnken design

pH and proportion of the buffer and wavelength of detection

Peak area and theoretical plate number

[38]

Sorafenib tosylate

RP-HPLC

ACN and water

65:35 v/v

Taguchi orthogonal arrays and Face centred cubic design

Mobile phase ratio and flow rate

Peak area, theoretical plates, retention time(Rt) and peak tailing

[39]

Tamoxifen

Citrate

ACN and phosphate buffer (pH 3.5) 52:48 v/v

Taguchi design and Box-Behnken design

Mobile phase ratio,

Buffer pH and oven temp

Peak area, retention time, theoretical plates, and peak tailing

[40]

Telmisartan and Hydrochlorothiazide

RP-HPLC

Mobile phase-A 0.02 M potassium

dihydrogen phosphate (pH of 3.5) and mobile phase-B- a mixture of Milli-Q water and

acetonitrile (100: 900 v/v) respectively

Three-Level Factorial design

Flow rate, column

Temperature and buffer pH

Resolution between drug and impurity

[41]

Fusidic acid (FA)

RP-HPLC

Methanol:

acetonitrile (5: 95, v/v)

Taguchi designand Central Composite Design

The ratio of solvents %w/w) and (Water %w/w)

Theoretical

Plates, assay

(%) and tailing

factor

[42]

Valsartan

RP-HPLC

Methanol, ACN, water, and buffers

Box–Behnken design

Mobile phase pH, flow rate, and % organic modifier

Peak area, retention time, theoretical plate count, and peak tailing (PT)

[43]

15 fixed-dose combinations (FDCs) of anti-hypertensive drugs

RP-HPLC

ACN-water (pH 6.2; 42:58 %, v/v).

Box-Behnken design

[44]

Rotigotine

RP-HPLC

ACN proportion: 54% v/v

Plackett-Burman design and Box-Behnken design

ACN proportion, pH of the buffer, and flow rate

The number of theoretical plates and retention time

[45]

Efavirenz

RP-HPLC

mobile phase: CAN 51.17%v/v

Plackett-Burman design and Box-Behnken design

ACN proportion, pH of the phosphate buffer, and mobile phase flow rate

Retention time and number of theoretical plates

[46]

Bosutinib

Stability-Indicating RP-HPLC Method

ACN-1.0% triethylamine (v/v) in water

Central composite design

Critical method attributes

Critical analytical attributes

[47]

ACN: Acetonitrile

Conclusion

In the pharmaceutical sector, AQbD is crucial for assuring method consistency and non-variability in outcomes. In order to improve quality, scientists can quickly identify the threads. The performance of analytical methods for currently available pharmaceuticals must be periodically reviewed to rectify any gaps and risk factors utilizing AQbD.

Acknowledgement

The authors are thankful to the management of Vels  Institute of Science, Technology and Advanced Studies (VISTAS), Pallavaram, Chennai-600 117, Tamil Nadu, India for providing Digital Library facility for completing this work successfully.

Conflict of Interest

The authors declare that there are no Conflicts of Interests among us.

 Funding Support

There are no funding Sources

References

  1. R. Phadke, A. Gosar, R. Mali and D. Patil,A Review on Quality by Design Approaches to Analytical Method Development. Indo Am. J. Pharm., 9, 2044 (2019).
    CrossRef
  2. S.B. Gholve, R.R. Ajgunde, O.G. Bhusnure and S.S. Thonte, Analytical Method Development and Validation by QbD Approach – A Review. Der Pharmacia Sinica, 6, 18 (2015).
  3. D. Taylor, The Pharmaceutical Industry and the Future of Drug Development, in Pharmaceuticals in the Environment, p. 1-33 (2015);
    CrossRef
  4. P. Ravisankar, S. Gowthamiand G. Devlala Rao, A Review on Analytical Method Development,IJRPB,2, 1183 (2014).
  5. T. Verch, C. Campa, C.C. Chéry, R. Frenkel, T. Graul, N. Jaya, B. Nakhle, J. Springall, J. Starkey, J. Wypych,  and T. Ranheim, Analytical Quality by Design, Life Cycle Management, and Method Control, The AAPS Journal,24,  34(2022)
    CrossRef
  6. L.X. Yu, G. Amidon, M. A. Khan, S.W. Hoag, J. Polli, G.K. Raju and J. Woodcock, Understanding Pharmaceutical Quality by Design,The AAPS Journal, 16,  771 (2014);https://doi.org/10.1208/s12248-014-9598-3
    CrossRef
  7. A.V. Ganorkar and K.R. Gupta, Analytical Quality by Design: A Mini Review, Biomed. J. Sci. Tech. Res.,1, 1555(2017).
    CrossRef
  8. R. Peraman, K. Bhadraya and Y.P. Reddy, Analytical Quality by Design: A Tool for Regulatory Flexibility and Robust Analytics,Int. J. Anal. Chem.,2015, 868727 (2015);
    CrossRef
  9. M. Schweitzer, M. Pohl, M. Hanna-Brown et al., Implications and Opportunities of Applying QbD Principles to Analytical Measurements, Pharm. Technol., 34, 52 (2010).
  10. P. Elder and P. Borman, Improving Analytical Method Reliability Across The Entire Product Lifecycle Using QbD Approaches, Pharm. Outsourcing, 14, 14(2013).
  11.  P. Borman, M. Chatfield, P. Nethercote, D. Thompson and K. Truman, The Application of Quality by Design to Analytical Methods, Pharm. Technol., 31, 142 (2007).
  12. U. S. Food and Drug Administration. Guidance for Industry: Q8(2) Pharmaceutical Development. 2009.
  13. A. Rajora and G. Chhabra, Quality by Design Approach: Regulatory Need, Current, and Future Perspective, Asian J. Pharm. Clin. Res., 14, 29 (2021).
    CrossRef
  14. Food and Drug Administration. Available from: http://www.fda.gov/ ohrms/dockets/ac/06/minutes/2006–4228m1.pdf.
  15. US Food and Drug Administration; Guidance for Industry: Q10 Pharmaceutical Quality Systems; 2009.
  16. M. Nasr. Risk-based CMC Review Paradigm, Advisory Committee for Pharmaceutical Science Meeting; 2004.
  17.  Food and Drug Administration CDER. Guidance for Industry: Immediate Release Solid Oral Dosage Forms Scale-up and Post approval changes: Chemistry, Manufacturing, and Controls, in vitro Dissolution Testing, and in vivo Bioequivalence Documentation; 1995, Volume 60, 61465-61644(1995).
  18. ASTM E2281—08a (2012)e1 Standard practice for process andmeasurement capability indices.http://www.astm.org/Standards/E2281.htm.
  19. J.A.De Feo and Barnard W, JURAN Institute’s six sigma breakthrough and beyond—quality performance breakthrough methods. India: Tata McGraw-Hill Publishing Company Limited; 2005
  20. Rajendra Phadke et al. A review on Quality by Design Approaches to Analytical Method Development, Indo American Journal of Pharmaceutical Research.2019:9(007)
  21. Patrick Jackson et al. Using the Analytical Target profile to Drive the Analytical Method Lifecycle, Anal. Chem.. 2009, 91, 4, 2577-2585(2019) 
    CrossRef
  22. M.Blessy, R.D. Patel, P.N. Prajapati and Y.K. Agrawal, Development of forced degradation and stability indicating studies of drugs—A review,J Pharma Analysis,4, 159 (2014)
    CrossRef
  23. B.R.Jena, S.P.Panda, U.Kulandaivelu, R.R.Alavala, G.S.N.K.Rao, S.Swain, G.Pattnaik and D.Ghose, AQbD Driven Development of an RP-HPLC Method for the Quantitation of Abiraterone Acetate for its Pharmaceutical Formulations in the Presence of Degradants,Turk. J. Pharm. Sci.,18,718 (2021);
    CrossRef
  24. S.Karmarkar, X.Yang, R.Garber, A.Szajkovics andM. Koberda, Quality by design (QbD) Based Development and Validation of an HPLC Method for Amiodarone Hydrochloride and its Impurities in the Drug Substance,J. Pharm. Biomed. Anal.,100, 167 (2014);
    CrossRef
  25. S.Jebali, C.Belgacem, M.R.Louhaichi, S.Bahri and L.Latrous El Atarche, Application of Factorial and Doehlert Designs for the Optimization of the Simultaneous Separation and Determination of Antimigraine Drugs in Pharmaceutical Formulations by RP-HPLC-UV,Int. J. Anal. Chem.,2019,9685750 (2019);
    CrossRef
  26. M.Yabré, L.Ferey, T.I.Somé, G.Sivadier andK.Gaudin, Development of a Green HPLC Method for the Analysis of Artesunate and Amodiaquine Impurities Using Quality by Design,J. Pharm. Biomed. Anal.,190, 113507 (2020);
    CrossRef
  27. N.K.Alruwaili, Analytical Quality by Design Approach of Reverse-Phase High-Performance Liquid Chromatography of Atorvastatin: Method Development, Optimization, Validation, and the Stability-Indicated Method,Int. J. Anal. Chem.,2021, 8833900 (2021);
    CrossRef
  28. R.Kaur, S.Saini, A.Patel, T.Sharma, R.Kaur, O.P.Katare and B.Singh, Developing a Validated HPLC Method for Quantification of Ceftazidime Employing Analytical Quality by Design and Monte Carlo Simulations,J AOAC Int.104, 620 (2021);
    CrossRef
  29. K.Y.Patel, Z.R.Dedania, R.R. Dedania and U. Patel,QbD Approach to HPLC Method Development and Validation of Ceftriaxone Sodium, Futur. J. Pharm. Sci.7, 141 (2021).
    CrossRef
  30. T.Tol, H.Tawde, S.Gorad, A.Jagdale, A.Kulkarni, A.Kasbale, A.Desai and G.Samanta, Optimization of A Liquid Chromatography Method for the Analysis of Related Substances in Daclatasvir Tablets Using Design of Experiments Integrated With the Steepest Ascent Method and Monte Carlo Simulation,J. Pharm. Biomed. Anal. 178 (2020) ;
    CrossRef
  31. V.C.Gurumukhi and S.B.Bari, Quantification and Validation of Stability-Indicating RP-HPLC Method for Efavirenz in Bulk and Tablet Dosage Form using Quality by Design (QbD): A Shifting Paradigm,J. Chromatogr. Sci.,60, 143 (2022);http://dx.doi.org/10.1093/ chromsci/bmab061
    CrossRef
  32. B.Jayagopal andS. Murugesh, QBD-driven HPLC Method of Eltrombopag Olamine: Degradation Pathway Proposal, Structure Elucidation, and In Silico Toxicity Prediction,J. Pharm. Biomed. Anal., 203, 114231 (2021);http://dx.doi.org/10.1016/j.jpba.2021.114231
    CrossRef
  33. R.Peraman, K.Bhadraya, Y.P.Reddy, C.S.Reddy and T.Lokesh Analytical Quality by Design Approach in RP-HPLC Method Development for the Assay of Etofenamate in Dosage Forms,Indian J. Pharm. Sci.77, 751 (2015);
    CrossRef
  34. S.Saini, T.Sharma, A.Patel, R.Kaur, S.K.Tripathi, O.P.Katare andB.Singh, QbD-Steered Development And Validation of An RP-HPLC Method for Quantification of Ferulic Acid: Rational Application of Chemometric Tools,J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.,1155, 122300 (2020);
    CrossRef
  35. N.K.Yadav, A.Raghuvanshi, G.Sharma, S.Beg, O.P.Katare andS. Nanda, QbD-Based Development and Validation of a Stability-Indicating HPLC Method for Estimating Ketoprofen in Bulk Drug and Proniosomal Vesicular System,J. Chromatogr Sci.,54, 377 (2016).
    CrossRef
  36. S.Beg, V.Chaudhary, G.Sharma, B.Garg, S.S.Panda and B.Singh, QbD-oriented Development and Validation of a Bioanalytical Method for Nevirapine With Enhanced Liquid-Liquid Extraction and Chromatographic Separation,Biomed. Chromatogr.,30, 818 (2016)
    CrossRef
  37. S.Beg, G.Sharma, O.P.Katare, S.Lohan and B.Singh, Development and Validation of a Stability-Indicating Liquid Chromatographic Method for Estimating Olmesartan Medoxomil Using Quality by Design,J. Chromatogr Sci.53, 1048 (2015);
    CrossRef
  38. A.V.Dalvi, C.T.Uppuluri, E.P.Bommireddy andP.R.Ravi, Design of Experiments-Based RP – HPLC Bioanalytical Method Development for Estimation of Rufinamide in Rat Plasma and Brain and its Application in Pharmacokinetic Study. J. Chromatogr. B Analyt. Technol. Biomed Life Sci.,1102-1103, 74 (2018);http://dx.doi.org/ 10.1016/j.jchromb.2018.10.014
    CrossRef
  39. T.Sharma, R.K.Khurana, A.Jain, O.P.Katare and B. Singh, Development of a Validated Liquid Chromatographic Method for Quantification of Sorafenib Tosylate in the Presence of Stress-Induced Degradation Products and in Biological Matrix Employing Analytical Quality by Design Approach, Biomed. Chromatogr.,32, e4169 (2018);
    CrossRef
  40. P.S.Sandhu, S.Beg, O.P.Katare and B.Singh, QbD-Driven Development and Validation of a HPLC Method for Estimation of Tamoxifen Citrate with Improved Performance,J. Chromatogr Sci.,54, 1373 (2016);http://dx.doi.org/10.1093/chromsci/bmw090
    CrossRef
  41. A.K.Palakurthi, T.Dongala and L.N.R.Katakam, QbD based development of HPLC method for simultaneous quantification of Telmisartan and Hydrochlorothiazide impurities in tablets dosage form,Pract. Lab Med.,21, e00169 (2020);
    CrossRef
  42. G.Sharma, K.Thakur, K.Raza and O.P.Katare, Stability Kinetics of Fusidic Acid: Development And Validation of Stability Indicating Analytical Method by Employing Analytical Quality by Design Approach in Medicinal Product(s),J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 1120, 113 (2019);
    CrossRef
  43. S.Bandopadhyay, S.Beg, O.P.Katare, T.Sharma andB.Singh, Integrated Analytical Quality by Design (AQbD) Approach for the Development and Validation of Bioanalytical Liquid Chromatography Method for Estimation of Valsartan, J. Chromatogr. Sci.,58,606 (2020);
    CrossRef
  44. P.B.Prajapati, A.S.Patel and S.A.Shah, DoE-Based Analytical-FMCEA for Enhanced AQbD Approach to MEER-RP-HPLC Method for Synchronous Estimation of 15 Anti-Hypertensive Pharmaceutical Dosage Forms,J AOAC Int.,105, 34 (2022);
    CrossRef
  45. P.Saha and M.M.Pandey, DoE-based Validation of a HPLC-UV Method For Quantification of Rotigotine Nanocrystals: Application to In vitro Dissolution and Ex vivo Nasal Permeation Studies,Electrophoresis,43, 590 (2022). http://dx.doi.org/10.1002/elps.202100157
    CrossRef
  46. P.Saha andM.M. Pandey, Design of Experiment (DoE)-Approach Based RP-HPLC Analytical Method Development and Validation for Estimation of Efavirenz in Bulk and Formulations,J. Chromatogr. Sci.,60, 35 (2022);
    CrossRef
  47. P.B.Prajapati, N.Bagul, G.Kalyankar, Implementation of DoE and Risk-Based Enhanced Analytical Quality by Design Approach to Stability-Indicating RP-HPLC Method for Stability Study of Bosutinib,J. AOAC Int.,104, 1742 (2021);
    CrossRef
  48. Kowtharapu, LP, Katari, NK, Sandoval, CA, Muchakayala, SK, Rekulapally, VK. Unique green chromatography method for the determination of serotonin receptor antagonist (Ondansetron hydrochloride) related substances in a liquid formulation, robustness by quality by design-based design of experiments approach. J Sep Sci. 2022; 45: 1711– 1726.
    CrossRef
  49. Kowtharapu, Leela Prasad and Katari, Naresh Kumar and Sandoval, Dr.Christian A. and Muchakayala, Mr.Siva Krishna and Rekulapally, Dr. Vijay Kumar, Green Liquid Chromatography Method for the Determination of Related Substances of Mast Cell Stabilizer and Histamine Receptor Antagonist in Nasal Spray Formulation, QbD Based Approach for Method Robustness Establishment. SSRN: https://ssrn.com/abstract=3992391.
    CrossRef
Share Button
(Visited 354 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.