View Article

Abstract

Quality by Design (QBD) is a systematic, statistical, structured, product development module commonly employed for the formulation development of drug products in pharmaceutical industry. The inherent purpose of QbD is to ensure consistent drug formulation quality, minimize risks, and enhance patient safety. This approach has now become almost indispensable to dosage form design and formulation optimization. The ability of QBD to delve into comprehensive understanding of drug product and its manufacturing process can make it a robust tool for the development of nanoformulations, whose scale-up is often limited by the numerous complexities involved in their development and optimization. Quality-by-Design can offer a structured framework facilitating swift optimization and simplistic scale-up. This review article offers technical insights into the significance of Quality-by-Design to ensure hassle-free development of innovative and effective nanotherapeutic products. Development of different types of nanoformulations aligned with the multi-step approach of QBD as outlined in the regulatory guidelines has been elaborated in the review. The article also briefly discusses Process Analytical Technology (PAT); another statistical-analytical based tool to facilitate better process control ensuring development of a robust product

Keywords

quality by design, critical factors, nanoformulation, nanotherapeutics, PAT, product design.

Introduction

Pharmaceutical product development not only implies development of a safe, efficacious, stable product that meets the needs of patients and delivers the expected performance; but also establishes a manufacturing process robust enough to consistently deliver a product of desired quality. Traditionally, a methodology known as ‘Quality by Testing’ was employed to ensure product quality. This approach heavily relied on intensive and repeated testing of raw materials, packaging materials, in-process quality control testing, and evaluation of final products. However, while this approach was singularly based on meeting product specifications, it seldom achieved complete product and process understanding and hence often failed to achieve its intended purpose1. Furthermore, an insufficient understanding of the product led to difficulties in identifying the root cause of formulation failures. Similarly, a lack of process comprehension prevented manufacturers from altering operating parameters without submitting change controls. Hence, a more progressive, risk-based approach supported by experimental design i.e. Quality by Design (QbD) evolved in the 90s decade that assured a quality product while also offering plenty of drug product and production process understanding2,3. The pharmaceutical arena has swiftly applied this strategy for the optimization and development of drug delivery systems, biotechnological products, and medical devices. While QbD has been successfully and consistently used in the preparation of conventional systems, the more complex web of design and development of nanoformulations can also be simplified by implementing QbD principles4,5.  This review discusses the principles of QbD and their applications in the optimization and development of nanotherapeutic dosage forms. The initial sections of the review delve into the origins and implications of QbD in the realm of pharmaceutical product development. This is followed by an exploration of the potential for applying QbD principles to the creation of nano-dosage forms. The latter part of the review demonstrates with suitable case-studies the applicability of QbD concepts to the preparation and optimization of a few prototype nanosystems, supported by examples from recent publications.

Quality by Design: Origins, Overview and Structure

Since its inception by Dr. Joseph Juran as a part of his quality trilogy in 1991, the concept of QbD has been extensively researched and adapted by numerous manufacturing units such as automobiles, electronics, and mass manufacturing general products6. Dr. Juran’s fundamental philosophy, which culminated into the conceptualization of QbD, was that ‘quality should be a deliberate choice and it should be incorporated within the product, rather than the notion of inspecting quality within the products’7,8. The QbD principle has since been implemented in numerous manufacturing companies to not only elevate their product quality, but also to simultaneously reduce associated costs 9,10. Quality by Design (QbD) has become a prerequisite for the pharmaceutical development industry, aiding in the creation of a diverse range of pharmaceutical, biotechnological products, and medical devices11,12. Being healthcare products, the quality of these preparations is of utmost significance. QbD is a reliable method to achieve the desired quality, eliminating the need for extended product review processes, preventing delays in market introductions, avoiding inconsistencies in product quality, and enhancing patient compliance13,14. QbD is integral to enhancing the optimization process for complex pharmaceuticals, as it systematically considers and evaluates various factors of variability that may influence the ultimate quality of the end product15. The development of QbD was backed by a number of quality guidelines published by the FDA and ICH; i.e. ICH Q8 (2004), ICH Q9 (2005) and ICH Q10 (2008) 16,17. ICH Q8 guideline outlines principles for pharmaceutical development to ensure that products meet predetermined quality criteria. The ICH Q9 guideline on the other hand offers directions on quality risk management principles and strategies relevant to pharmaceutical development, manufacturing, and quality control processes18,19. The ICH Q10 guideline provides a thorough framework for pharmaceutical quality systems, emphasizing a holistic approach to ensure consistent product quality and continual improvement throughout the product lifecycle20. QbD is thus a new ray of hope for formulators to accelerate the formulation and optimization of drug products with lower instances of rejected drug products and lower development costs21–23.

Nanoformulations: Types and Characteristics

Nanomedicine have achieved burgeoning importance in the area of drug delivery systems and clinical therapy; due to its ability to counter the demerits of conventional dosage forms and older drug delivery systems such as frequent dosage regimen, requirement of high doses, lack of target specificity, and side effects24–27. Nanoformulations consist of drug molecules encapsulated within a lipidic or polymeric coat or drug dispersed within a matrix of the lipid or polymer with their size in the range of 10-1000 nm28. Nanomedicines offer numerous advantages to drug release like increased surface area, higher solubility, targeted drug release, better permeation, higher bioavailability, and amenability to the attachment of numerous ligands thereby facilitating active targeting29. A few commonly researched nanosystems include liposomes, niosomes, dendrimers, polymeric nanoparticles, lipidic nanoparticles, metallic nanoparticles, nanocrystals, etc30. Some recently discovered nanosystems such as bilosomes, silica-based systems, fullerenes are also attracting interest from researchers. More than 1,20,000 scientific articles have been published in the last few years exploring applications of nanosystems to various conditions such as infectious diseases, tumor therapy, and CNS delivery. Several marketed products such as Emend (Aprepitant), TriCor (Fenofibrate), Myocet (Doxorubicin), Abraxane (Paclitaxel), Rapamune (Sirolimus), Ambisome (Amphotericin B), etc. developed as a nanosystem have been commercialized recently31–33.

Scope of QbD in the Design and Commercialization of Nanotherapeutics

The preceding section markedly emphasizes the wide array of applications of nanoformulations to accentuate the therapeutic efficacy of drug molecules. However, the pace of commercialization of nanoformulations does not match the extravagant pace of research in this area34. Some of the glaring challenges faced by formulators in this area include: i. nanoformulations involve multiple and varied categories of excipients and multivariate manufacturing processes that vary depending upon the type of nanoformulation; ii. exquisite understanding of the impact of such a diverse range of excipients and manufacturing steps is often unfathomable iii. lack of clarity in understanding the CQAs of such formulations; iv complexity in the manufacturing process and process design; v. low reproducibility and extensive variability in the manufacturing outcomes; vi difficulties in scale-up of the sophisticated manufacturing processes involved; vii concerns about the toxicity potential of the products and by-products; and viii. safety and therapeutic efficacy-related regulatory challenges35,36. A lack of understanding of the impact of manufacturing steps and other formulation variables drastically hamper product quality. Negligence in monitoring product quality may not only impact the business prospects but also result in severe consequences to the patient culminating in legal consequences37,38.  An uncomplicated approach to sort these challenges would be an exhaustive understanding of the role and functionality of the formulation components and processing steps, and studying their influence individually and in combination with the critical product characteristics at the very beginning of the formulation development step itself. Contrary to the conventional approaches, the systematic QbD approach driven by analytical and risk-management methodologies links the desired attributes in a product with the formulation and manufacturing variables early in development39. Risk assessment strategies, software-assisted statistics, and data analytics enable formulators to fully comprehend if and how these variables affect product characteristics. Critical product characteristics for nanoformulations include particle size and polydispersity index, zeta potential, encapsulation efficiency and drug loading, in vitro drug release, particle morphology, drug disposition profile, stability, and impurity profile to name a few40.  A robust control strategy in correspondence with PAT sensors and analyzers assures control of the highly significant variables, thereby ensuring consistency in product quality41,42.

Development of Nanopharmaceuticals based on QbD

ICH Q8 (R2) guideline defines QbD as an approach to pharmaceutical development that emphasizes understanding and controlling the manufacturing process to ensure the final product meets predefined quality standards. QbD is executed via the following five steps:

  • Drafting a Target Product Profile (TPP)
  • Critical Quality Attributes (CQAs)
  • Identifying Critical Material Attributes (CMAs) and Critical Process Parameters (CPPs)
  • Risk Assessment linking the CMAs and CPPs to CQAs
  • Design Space and Control Strategy29

The following sections detail the execution of these steps for the development of nanoformulations with emphasis on specific prototype nanosystems such as Liposomes, Niosomes, Polymeric and Lipidic Nanoparticles, Nanocrystals, and Micelles. Liposomes and Niosomes are self-assembling vesicles comprising of phospholipids and non-ionic surfactants respectively43. These vesicles are extensively researched owing to their ability to encapsulate drugs with both hydrophilic and lipophilic functional groups and prolonged circulation time44.  Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) represent the two generations of lipid-based nanosystems that have been extensively researched for achieving drug targeting. Lipid-based nanoformulations are appealing due to their biological similarities and a multitude of benefits. These include their capability to carry both water-loving (hydrophilic) and fat-loving (lipophilic) drugs, enhanced solubility of drugs and their permeation through biological barriers, their ability to respond to stimuli, as well as their compatibility and degradability in biological systems. These nanosystems also have the ability to protect their contents, which prolongs the drug’s half-life in the bloodstream and prevents absorption by immune cells, thereby reducing the risk of systemic toxicity45–47. On similar lines, polymeric nanoparticles are composed of biodegradable polymers that maybe of natural or synthetic origin, used for active and passive targeting of numerous drug molecules48. Nanocrystals are a relatively newer category of carrier-free nanosystems which constitute of only nanosized drug crystals stabilized with an optimum surfactant that confers stearic and thermodynamic stability to the system . The average particle size of nanocrystals ranges from 10 – 1000 nm49,50. Micellar structures are generated when surfactants are dissolved in an aqueous liquid; mostly water and entrap hydrophobic drugs. Often a polymer may also be incorporated in the micelles, where the system is referred to as ‘polymeric micelles’51.

A. Drafting a Target Product Profile (TPP): A TPP implies the characteristic features desired in the intended product based on the available literature and past experiences. The TPP is a living depiction of the product, which needs to be regularly updated throughout the product’s lifecycle. The Quality Target Product Profile (QTPP) is created based on the TPP as a blueprint, and it outlines the goals for product development. A thorough QTPP should encompass the properties of the active pharmaceutical ingredient, its functional traits, and the clinical ramifications of the final product for the specified patient population52. Critical Quality Attributes (CQAs) are crucial properties of a pharmaceutical product that are identified from the QTPPs based on whether they influence the safety, effectiveness of the product, and degree of the harm it may cause to the patient.

Being similar in functional properties, the QTPPs and CQAs for most of the nanosystems overlap with each other as mentioned in Table I. It is important to note here that the enlisted QTPPs and CQAs are based only on general examples and may deviate depending upon the route of administration. In specific cases i.e. a nanosystem to be delivered by the ocular/ parenteral route will also include sterility, isotonicity, and viscosity as the QTPPs and CQAs. Further, nanosystems delivered by the inhalation route will include aerodynamic diameter as one of the QTPP and CQA53,54 .

 


Table I: QTPPs and CQAs for Nanoformulations

 

Type of Nanosystem

QTPPs

CQAs

Liposomes

Indication, type of dosage form, route of administration, physical and chemical stability, liposomal size and polydispersity, liposomal surface charge and morphology, assay, encapsulation efficacy and drug release39,55,56

Particle size, surface charge, morphology, physical stability, and drug release, entrapment efficiency39,55,56

Niosomes

Same as that for liposomes57

Polymeric Nanoparticles

Particle size and polydispersity, morphology, surface charge, drug loading capacity, and drug release kinetics. Stability, encapsulation efficiency, biocompatibility, long-term stability58–60

Particle size, surface charge, morphology, physical stability, and drug release, entrapment efficiency58–60

Lipidic Nanoparticles

Particle Size and polydispersity, morphology, surface charge, drug loading capacity, and drug release kinetics. Stability, encapsulation efficiency, biocompatibility, long-term stability61–63

Particle size, surface charge, morphology, physical stability, and drug release, entrapment efficiency61–63

Nanocrystals

Type of dosage form, route of administration, particle size and polydispersity, physical stability31,64

Particle size and polydispersity, zeta potential, morphology, solid-state form, and physical stability31,64

Polymeric Micelles

Indication, type of dosage form, patient population, Administration route, Site of activity, Dosage strength, Dosage form, Viscosity, Osmolality, pH, Mucoadhesive properties (depending upon indication and route of administration), Particle characteristics (Particle Size and polydispersity, morphology, surface charge, drug loading capacity, drug release kinetics. Stability, encapsulation efficiency), Safety, Solubility, Drug release, Drug permeability65,66

Particle size, surface charge, morphology, physical stability, and drug release, entrapment efficiency65,66


B. Identifying CMAs and CPPs: CMAs are those properties of the raw materials i.e. API and excipients that have a markedly significant effect on the drug product CQAs. A typical CMA maybe the concentration of an API or excipient or their bulk characteristics like particle size, polymorphic nature, presence of pseudopolymorphs, or molecular weight/ viscosity of the polymer. Likewise, CPPs constitute those parameters of the unit operations performed while preparing a formulation that affects the product quality. This may include parameters like the speed of mixing during a process, processing time and temperature, amount of pressure applied, pH, etc. Due to the varied number of excipients and numerous methods of preparation involved, the CMAs and CPPs for the development of nanosystems distinctly vary from each other67. Table II depicts the CMAs for different nanoformulations, while Table III represents the CPPs involved depending upon the method of preparation.

 


Table II: CMAs to be Studied for Nanoformulations

 

Type of Nanosystem

CMAs

Liposomes

Phospholipid: cholesterol molar ratio, Drug:lipid ratio, Type and conc. of cryoprotectant, Phase transition temperature of the lipid37

Niosomes

Drug concentration, Surfactant: cholesterol ratio, Type, and conc. of cryoprotectant68

Polymeric Nanoparticles

Type and concentration of polymer, Drug: polymer ratio, Polymer: surfactant ratio, Solvent type, and composition, Polymer molecular weight, Salting out agent concentration69,70

Lipidic Nanoparticles

Drug concentration, Type and concentration of the lipid, Drug: lipid concentration, Lipid: surfactant concentration71,72

Nanocrystals

Type of surfactant, Drug loading, Drug: surfactant ratio31

Polymeric Micelles

Polymer properties such as molecular weight, log P, CMC value, HLB value, and temperature concentration solubility, LD50 value; drug properties such as molecular weight, log P, melting point, solubility, LD50 value, solvent properties including pH, ionic strength, volume, temperature; excipient properties including those of surfactants and cryoprotectants65,66


Table III: CPPs to be Studied for the Various Preparation Methods of Nanoformulations

 

Type of Nanosystem

Method of Preparation

CPPs

Liposomes73

Thin Film Hydration

Speed of rotation, Hydration time, Hydration temperature, pH of the buffer

Emulsification solvent removal

Speed and time of agitation, Speed of mixing of phases, Temperature of mixing, Injection speed

Sonication

Sonication time

Extrusion

Extrusion temperature, No. of passes through the extrusion membrane

Freeze Thawing

Freezing temperature

Niosomes70

Same as that for liposomes

Polymeric Nanoparticles74

Solvent evaporation

Mixing speed and time, evaporation rate, temperature and humidity, sonication time and amplitude, drying time, and temperature

Emulsification/ solvent diffusion

Emulsification parameters (high-pressure homogenization, ultrasonication), solvent diffusion rate, temperature, and cooling rate

Reverse Salting Out

Mixing time, and temperature, and drying time, and temperature

Nanoprecipitation

Mixing speed, anti-solvent addition rate, temperature, pH, drying parameters (freeze-drying or spray-drying

Lipidic Nanoparticles75

Homogenization

Homogenization speed, Homogenization pressure, No. of passes through the homogenizer

Sonication

Sonication time, Amplitude of sonication

Microfluidization

Microfluidization pressure, No. of passes through the microfluidizer

Nanocrystals31

Media Milling

Weight of milling media, Milling speed, Milling time

Polymeric Micelles76

Direct dissolution

Solubilization of API, solubilization of the polymer, mixing time, rotation speed, sequence of addition of excipients

 

Dialysis method

Diameter of the dialysis tube, flow rate and dialysis time, solubility, and contact volume.

 

Oil-in-water emulsion method

Time and temperature of mixing and addition, Phase separation

 

Freeze drying method

Freezing time and temperature; Time, pressure, and pressure of drying; Vapor pressure

 

Thin film method/ vacuum evaporation

Temperature, speed, and duration of rotation, hydration time and properties of hydration media,  starting and ending pressure, scale of decompression,


C. Risk Assessment linking the CMAs and CPPs to CQAs: Risk assessment as per either of the quality risk management (QRM) tools enlisted in the guideline ICH Q9 is performed to link the CMAs and CPPs to CQAs19. Risk estimation matrix (REM) and failure mode effect analysis (FMEA) are among the frequently utilized tools in Quality Risk Management (QRM)77. On the other hand, Ishikawa fish-bone diagrams are typically chosen for establishing cause-and-effect connections. Risk assessment allows the identification of extremely crucial CMAs and CPPs which are to be further optimized using statistical-based experimental designs78,79.

The example given below in Figure 1 relates to the development of PLGA nanoparticles by a modified nanoprecipitation method80. The fishbone diagram illustrates how the variables of formulation and process impact the quality of nanoparticle products. The diagram enables the formulator to visualize and identify the CMAs and CPPs that may be deemed as high risk.

       
            Ishikawa Fishbone Diagram for PLGA Nanoparticles Prepared by Modified Nanoprecipitation Method.png
       

Figure 1: Ishikawa Fishbone Diagram for PLGA Nanoparticles Prepared by Modified Nanoprecipitation Method

Table IV represents a Risk Estimation Matrix to perform a risk assessment of a thermosensitive liposome formulation composed of a blend of phospholipids prepared by thin film hydration followed by vacuum drying. The CMAs/ CPPs are designated as High/ Medium/ Low risk depending upon the possibility of the risk occurring and the severity of the consequences. The parameters deemed high risk i.e. the amount of phospholipids incorporated in the formulation were further optimized using statistical-based optimization tools39.


Table IV: Risk Estimation Matrix for Liposomal Formulation Prepared by Thin Film Hydration Method

 

Process

Composition

Dissolution of Lipids

Rotary Evaporation

Hydration Phase

Stabilization

Vacuum Drying

CPP/CMA

 

CQA

 

 

Amount of DPPC

Amount of DSCP

Amount of DSCPE-PEG30000

Solvent type

Solvent pH

Rotary evaporator (temperature, pressure)

Time

Temperature

Type of Cryoprotectant

Cryoprotectant  concentration

Membrane pore size

Particle size

H

H

H

M

M

M

M

L

M

L

M

Particle size distribution

M

M

M

L

L

L

M

L

M

M

M

Zeta potential

M

M

M

L

L

M

L

L

M

M

L

Morphology

L

L

L

M

M

L

M

L

L

M

M

Phase transition temperature

H

H

M

L

L

L

M

L

M

L

L


In another example of risk assessment as presented in Table V, FMEA was used for the risk evaluation of venlaflaxine-loaded lipidic nanocarriers by high-pressure homogenization method81. Risk parameters were evaluated quantitatively, taking into account the severity of the risk, the likelihood of its occurrence, and the simplicity of its detection. Those risk parameters that had a higher ‘Risk Priority Number’ (RPN), which is determined as the product of the three scores, were subjected to further optimization and stringent control.


Table V: Failure Mode Effect Analysis of a Lipidic Nanocarrier Formulation by High Pressure Homogenization Method

 

Process parameter component

Failure mode

Failure effect

S

Potential cause of root of failure

O

Detectability method or control

D

RPN (S*O*D)

Mutual miscibility of the solid and liquid lipid

Inadequate proportion of solid to liquid lipid

Leaching out of drug, Phase separation

5

Inadequate quantity and formulation composition

2

Visual observation, DSC analysis

2

20

Drug: solid + lipid ratio (D:L)

Insufficient quantity of the lipids

Insufficient drug loading, poor stability

5

Insufficient amount of liquid lipid

5

?

2

50

Conc. of Surfactant

Inadequate amount

Improper size, PDI, and ?

5

Inadequate surfactant concentration

5

Particle size

2

50

Speed and time of high speed homogenizer

Incorrect homogenization process

Incorrect size, and entrapment,  Phase separation

4

Incorrect homogenization process

4

Particle size

2

32


D. Statistical-Based Optimization: Following the risk assessment, the screened high-risk parameters are further optimized using software-assisted optimization designs, generically referred to as ‘Design of Experiments (DoE)’. DoE is an efficient tool that eliminates the wastage of time and money due to the conventional trial and error methodologies that involve exhaustive experimentation with no assurance of a quality product.  Design of Experiments (DoE) provides an enhanced comprehension of the complex interplay of dependent and independent variables in formulation development82. These experimental designs fall into categories known as screening designs (that are used to classify the more important factors from the lesser ones when an extremely large number of factors are to be studied) and optimization designs (used to obtain the design space i.e. an optimized formulation). Screening designs commonly used include Plackett-Burman design (PBD), fractional factorial design (FFD), and Taguchi design (TD which can screen up to 31, 21, and 63 factors respectively. Optimization is performed using response surface designs (RSD) that are capable of quantifying the relationship between multiple variables and one or more responses75,83. The more popularly used RSD for optimization is the central composite design (CCD) capable of studying 5 levels for each factor and the box-behnken design (BBD) which studies every factor at 3 levels and hence generates a lesser number of trials compared to the CCD. For screening variables, a two-level factorial or fractional factorial design can be employed, whereas an optimization process can utilize a high-resolution factorial design. Both full factorial and fractional design methods allow for the examination of numerous factors by assigning two levels to each - a higher and a lower one84,85 . Other designs commonly employed include D-optimal design (D-OD) and mixture design (MD)86,87. Table VI depicts numerous examples of nano-dosage forms with the experimental designs used for optimization.

 


Table VI: Optimization of Nanoformulations by Statistical Optimization Methods

 

Type of Nanoformulation

Drug Incorporated

Screening/ Optimization Design Used

References

Liposomes

Cefoperazone

BBD

88

 

Simvastatin

CCD

56

 

Prednisolone

D-OD

37

 

Pravastatin

D-OD

55

 

Azacitidine

BBD

72

Niosomes

Casiopeina

Plackett-Burman, CCD

52

 

Lacidipine

BBD

44

 

Polymyxin

BBD

89

 

Levosulpiride

BBD

70

 

Dynorphin-B

BBD

57

Polymeric Nanoparticles

Cinacalcet HCl

Taguchi, BBD

69

 

Flurbiprofen

Plackett-Burman, CCD

58

 

Pioglitazone

3-level full factorial design

59

 

Quercetin

FFD with Resolution IV; 6 variables studied at 2 levels

90

 

Berberine chloride

32 Full Factorial Design

60

Lipidic Nanoparticles

Rivastigmine

BBD

91

 

Fluoxetine HCl

32 Full Factorial Design

62

 

Phenobarbital

Fractional Factorial, CCD

92

 

Atazanavir

CCD

63

 

Resveratrol

Plackett-Burman, BBD

54

Polymeric Micelles

Posaconazole

32 Full Factorial Design

93

 

Resveratrol

CCD

65

 

Galantamine

Taguchi, CCD

66


Executing the batches recommended by the software in experiments, along with analyzing the results through various plots and charts generated by statistical analysis, enhances cognizance of both the product and the process. The visual representation illustrates the influence of individual factors.

E. Design Space and Control Strategy

ICH Q8 (R2) defines design space as the multifaceted combination and interplay of dependent variables i.e. formulation contents and process parameters that have been proven to assure quality18. Simply put, design space is the mutual effect of CQAs, CMAs, and CPPs within which the product falls within the desired specifications of the QTPP. The design space; a segment-criteria the experimental realm, ensures that the pharmaceutical dosage form meets all the criteria of safety, identity, purity, and strength. So long as alterations made to the manufacturing process fall within the designated design space, they are not deemed as changes. Any adjustments outside the design space necessitate a post-approval change process following regulatory approval94. The control strategy is established as a mandatory regulatory requirement at the conclusion of the Quality by Design (QbD) based product development process. It serves as a set of critical factors influencing the variability in Critical Quality Attributes (CQAs) and process controls, ensuring ongoing consistency in the quality of the final product. Essential input parameters identified during the product development phase are documented as quality control strategies and continuously monitored throughout each batch operation4,17. Narayan et al. formulated nanocrystals of aceclofenac by wet-milling approach by adapting QbD methodology. Statistical optimization was performed using BBD to optimize four CMAs/CPPs i.e. size of milling media (5-15 mm), concentration of stabilizer (0.25-0.75 %), drug loading (200-400 mg), and milling time (1-4 hours). The four factors were evaluated at three levels to optimize the particle size, PDI, and surface charge of the system. Aceclofenac nanocrystals manufactured by loading 200 mg of the drug in 0.25% of the stabilizer solution for 4 hours using zirconium beads of size 5mm yielded nanocrystals as per the desired QTPP. These optimized parameters represent the design space64.

PAT for Optimization of Nanotherapeutics

The evolution of the Process Analytical Technology (PAT) concept was driven by the formulator's necessity to institute a science-based methodology for process control. This facilitates the dynamic management of the production and mitigates patient risks in the final dosage form. In direct terms, PAT allows control of CQAs by continuous monitoring of the CPPs and CMAs. In line with this, ICH Q8 emphasizes the adoption of PAT to uphold ensure processes operate within predefined design spaces95. PAT involves a range of methods that encompass techniques such as collecting data, analyzing multiple variables, controlling processes, continuously improving, and utilizing existing scientific expertise. This is achieved through at-line, on-line, and in-line monitoring and control of the process 62,61. Besides enhancing the quality and consistency of the product, PAT can contribute to heightened first-pass outputs, surplus management, lower batch failures, and lower production cycling time. PAT plays a pivotal role in pharmaceutical continuous manufacturing processes, where real-time process control is immensely crucial. Moreover, it accelerates the implementation of real-time release testing96,97. Table VII presents a list of commonly used spectroscopic techniques as PAT processes with a brief note on their common applications in pharmaceutical process monitoring.

 


Table VII: List of PAT Techniques98,99

 

PAT Process

Application

Near Infrared spectroscopy (NIR)

% Drug content or % Water content in powder mixing and drying processes

Tunable Diode Laser Spectroscopy (TDLS)

Headspace analysis, Lyophilisation

UV-Visible spectroscopy (UV Vis)

Used in hot melt extrusion for analysis of residence time. Blend uniformity, presence of amorphous vs. crystalline phases

Raman Spectroscopy

Determination of chemical structure, identification of polymorphs

Focused Beam Reflectance Measurements (FBRM)

Particle size, particle growth, size, and surface area

X Ray Fluorescence

Elemental analysis


A host of newer tools are used monitoring the CQAs involved in the manufacturing of nanoformulations i.e. particle size and polydispersity. Dynamic Light Scattering (DLS) microscopy based on Spectral Domain Optical Coherence Tomography (SDOCT) and Multi-angle static light scattering are used in-line monitoring of particle size. DLS which is used as the basis for analysis of particle size distribution in a flowing sample; can identify and analysing even dense concentrations without dilution. SDOCT on the other hand can collect data for nanoparticle samples at variable depths. Such tools broaden the horizons of PAT application to the process monitoring of nanoformulations by facilitating data regarding particle dimensions and flow characteristics. A groundbreaking non-destructive PAT tool, known as the 'NanoFlowSizer,' is currently in the developmental pipeline. Functioning on the axiom of Spectral-Domain Low-Coherence Interferometry, this tool offers continuous and in-line characterization of particle size and polydispersity for stationary as well flowing samples of nanoformulations. The device offers path length resolution along with dynamic light scattering data which also accounts for the influence of flow and/or multiple scattering100. Another novel PAT based tool is the Focused beam reflectance measurement for surveilling the appearance, concentration, and structural configuration of particles based on the statistical distribution of chord lengths drawn across the particles within a given dataset or system. This chord length distribution help in characterizing the size and distribution of the particles within the sample. ParticleTrack and EasyViewer probes working on this principle efficiently distinguish between a stable nanodispersion and one containing agglomerates. They have also been applied for the detection of small deviations in the range of pore sizes within a material such as an adsorbent as it interacts with an antigenic adjuvant. The pore size distribution is crucial because it determines if the antigenic adjuvant can penetrate and retain within the adsorbent.  To synopsize, PAT tools may exert a significant influence in on-line and/or at-line evaluation of nanoformulations as well, ensuring a quality product as per the principles of QbD. When complemented with digitized approaches and computerized methodologies, their impact would escalate exponentially leading to enhanced digital production and data analytics101–103.

Closing Remarks:

The pivotal phase in the approval process of a drug product involves ensuring that the formulator adheres to the specifications set by the regulatory agency. An elaborate examination and comprehension of the CMC of the active ingredient, excipient profiles, and multivariate production steps is crucial for the development of a stable, and quality nanoformulation. The presence of numerous factors and variables affecting the final product further introduces complexities in achieving reproducible product quality. Thus, Quality by Design with its principle of achieving consistent product performance with end goals in mind can serve as a fine tool to accelerate the commercialization of nanoformulations while saving crucial resources. Though regulatory requirements do not mandate the adoption of such paradigms, QbD-based development is certainly encouraged as is evident by the recent news articles and white papers, which emphasize that regulatory agencies prefer applications with QbD-based development.

REFERENCES

  1. Yu LX, Amidon G, Khan MA, Hoag SW, Polli J, Raju GK, Woodcock J. Understanding pharmaceutical quality by design. The AAPS journal. 2014 Jul;16:771-83.
  2. Zhang L, Mao S. Application of quality by design in the current drug development. Asian journal of pharmaceutical sciences. 2017 Jan 1;12(1):1-8.
  3. Thomson DS, Austin SA, Devine-Wright H, Mills GR. Managing value and quality in design. Building Research & Information. 2003 Jan 1;31(5):334-45.
  4. Beg S, Rahman M, Kohli K. Quality-by-design approach as a systematic tool for the development of nanopharmaceutical products. Drug discovery today. 2019 Mar 1;24(3):717-25.
  5. Sangshetti JN, Deshpande M, Zaheer Z, Shinde DB, Arote R. Quality by design approach: Regulatory need. Arabian Journal of chemistry. 2017 May 1;10:S3412-25.
  6. Bhoop BS. Quality by Design (QbD) for holistic pharma excellence and regulatory compliance. Pharm times. 2014;46(8):26-33.
  7. Juran JM. Juran on quality by design: The new steps for planning quality into goods and services. Simon & Schuster; 1992.
  8. Bala I, Verma S, Namdev KK. A Review on Process Variables Effects of an Oral Dosage Form by Using “Quality by Design”. International Journal of Drug Delivery Technology. 2015;5(1).
  9. Lionberger RA, Lee SL, Lee L, Raw A, Yu LX. Quality by design: concepts for ANDAs. The AAPS journal. 2008 Jun;10:268-76.
  10. Jeevanandam J, San Chan Y, Danquah MK. Nano-formulations of drugs: recent developments, impact and challenges. Biochimie. 2016 Sep 1;128:99-112.
  11. Rathore AS, Winkle H. Quality by design for biopharmaceuticals. Nature biotechnology. 2009 Jan;27(1):26-34.
  12. Borman P, Chatfield M, Nethercote P, Thompson D, Truman K. The application of quality by design to analytical methods. Pharmaceutical Technology. 2007 Oct 1;31(10):142-8.
  13. Javed MN, Alam MS, Waziri A, Pottoo FH, Yadav AK, Hasnain MS, Almalki FA. QbD applications for the development of nanopharmaceutical products. InPharmaceutical quality by design 2019 Jan 1 (pp. 229-253). Academic Press.
  14. Tefas LR, RUS L, Achim M, Vlase L, TOMU?? I. APPLICATION OF THE QUALITY BY DESIGN CONCEPT IN THE DEVELOPMENT OF QUERCETIN-LOADED POLYMERIC NANOPARTICLES. Farmacia. 2018 Sep 1;66(5).
  15. Patil AS, Pethe AM. Quality by Design (QbD): A new concept for development of quality pharmaceuticals. International journal of pharmaceutical quality assurance. 2013 Apr;4(2):13-9.
  16. Yu LX. Pharmaceutical quality by design: product and process development, understanding, and control. Pharmaceutical research. 2008 Apr;25:781-91.
  17. Nadpara NP, Thumar RV, Kalola VN, Patel PB. Quality by design (QBD): A complete review. Int J Pharm Sci Rev Res. 2012;17(2):20-8.
  18. ICH Harmonised Tripartite Guideline Q8 (R2) on Pharmaceutical Development. Available at: https://database.ich.org/sites/default/files/Q8_R2_Guideline.pdf. Accessed January 2024.
  19. ICH Harmonised Tripartite Guideline Q9 on Quality Risk Management. Available at: https://database.ich.org/sites/default/files/Q9_Guideline.pdf. Accessed January 2024.
  20. Reddy VV, Vishal Gupta N, Raghunandan HV, Nitin Kashyap U. Quality risk management in pharmaceutical industry: a review. International Journal of Pharm Tech Research. 2014;6(3):908-14
  21. Hasnain MS, Siddiqui S, Rao S, Mohanty P, Jahan Ara T, Beg S. QbD-driven development and validation of a bioanalytical LC–MS method for quantification of fluoxetine in human plasma. Journal of Chromatographic Science. 2016 May 1;54(5):736-43.
  22. Short SM, Cogdill RP, DRENNEN III JK, ANDERSON CA. A New Definition of Pharmaceutical Quality: Assembly of a Risk Simulation Platform to Investigate the Impact of Manufacturing/Product Variabcpility on Clinical Performance. Journal of pharmaceutical sciences. 2010 Dec 1;99(12):5046-59.
  23. Haleem RM, Salem MY, Fatahallah FA, Abdelfattah LE. Quality in the pharmaceutical industry–A literature review. Saudi pharmaceutical journal. 2015 Oct 1;23(5):463-9.
  24. Madaan T, Pandey S, Talegaonkar S, Delhi N. Nanotechnology: A smart drug delivery tool in modern healthcare. Journal of Chemical and Pharmaceutical Research. 2015;7(6):257-64.
  25. Khiev D, Mohamed ZA, Vichare R, Paulson R, Bhatia S, Mohapatra S, Lobo GP, Valapala M, Kerur N, Passaglia CL, Mohapatra SS. Emerging nano-formulations and nanomedicines applications for ocular drug delivery. Nanomaterials. 2021 Jan 12;11(1):173.
  26. Zhou X, Hao Y, Yuan L, Pradhan S, Shrestha K, Pradhan O, Liu H, Li W. Nano-formulations for transdermal drug delivery: A review. Chinese Chemical Letters. 2018 Dec 1;29(12):1713-24.
  27. Pathak K, Raghuvanshi S. Oral bioavailability: issues and solutions via nanoformulations. Clinical pharmacokinetics. 2015 Apr;54:325-57.
  28. Jiang W, Kim S, Zhang X, Lionberger RA, Davit BM, Conner DP, Lawrence XY. The role of predictive biopharmaceutical modeling and simulation in drug development and regulatory evaluation. International journal of pharmaceutics. 2011 Oct 14;418(2):151-60.
  29. Farjadian F, Ghasemi A, Gohari O, Roointan A, Karimi M, Hamblin MR. Nanopharmaceuticals and nanomedicines currently on the market: challenges and opportunities. Nanomedicine. 2019 Jan 1;14(1):93-126.
  30. Patra JK, Das G, Fraceto LF, Campos EV, Rodriguez-Torres MD, Acosta-Torres LS, Diaz-Torres LA, Grillo R, Swamy MK, Sharma S, Habtemariam S. Nano based drug delivery systems: recent developments and future prospects. Journal of nanobiotechnology. 2018 Dec;16:1-33.
  31. Peltonen L. Design space and QbD approach for production of drug nanocrystals by wet media milling techniques. Pharmaceutics. 2018 Jul 25;10(3):104.
  32. Ragelle H, Danhier F, Préat V, Langer R, Anderson DG. Nanoparticle-based drug delivery systems: a commercial and regulatory outlook as the field matures. Expert opinion on drug delivery. 2017 Jul 3;14(7):851-64.                 
  33. Chavda VP. Nanobased nano drug delivery: a comprehensive review. Applications of targeted nano drugs and delivery systems. 2019 Jan 1:69-92.
  34. Birla D, Khandale N, Bashir B, ShahbazAlam M, Vishwas S, Gupta G, Dureja H, Kumbhar PS, Disouza J, Patravale V, Veiga F. Application of quality by design in optimization of nanoformulations: Principle, perspectives and practices. Drug Delivery and Translational Research. 2024 Aug 10:1-33.
  35. Zieli?ska A, Carreiró F, Oliveira AM, Neves A, Pires B, Venkatesh DN, Durazzo A, Lucarini M, Eder P, Silva AM, Santini A. Polymeric nanoparticles: production, characterization, toxicology and ecotoxicology. Molecules. 2020 Aug 15;25(16):3731.
  36. Singh B, Sharma T, Saini S, Kaur R, Jain A, Raza K, Beg S. Systematic development of drug nanocargos using formulation by design (FbD): an updated overview. Critical Reviews™ in Therapeutic Drug Carrier Systems. 2020;37(3).
  37. Sylvester B, Porfire A, Muntean DM, Vlase L, Lupu? L, Licarete E, Sesarman A, Alupei MC, Banciu M, Achim M, Tomu?? I. Optimization of prednisolone-loaded long-circulating liposomes via application of Quality by Design (QbD) approach. Journal of liposome research. 2018 Jan 2;28(1):49-61.
  38. Simoes A, Veiga F, Figueiras A, Vitorino C. A practical framework for implementing Quality by Design to the development of topical drug products: Nanosystem-based dosage forms. International journal of pharmaceutics. 2018 Sep 5;548(1):385-99.
  39. Dobó DG, Németh Z, Sipos B, Cseh M, Pallagi E, Berkesi D, Kozma G, Kónya Z, Csóka I. Pharmaceutical development and design of thermosensitive liposomes based on the QbD approach. Molecules. 2022 Feb 24;27(5):1536.
  40. Charoo NA, Shamsher AA, Zidan AS, Rahman Z. Quality by design approach for formulation development: a case study of dispersible tablets. International journal of pharmaceutics. 2012 Feb 28;423(2):167-78.
  41. Bartelds R, Nematollahi MH, Pols T, Stuart MC, Pardakhty A, Asadikaram G, Poolman B. Niosomes, an alternative for liposomal delivery. PLoS One. 2018 Apr 12;13(4):e0194179.
  42. Zagalo DM, Silva BM, Silva C, Simoes S, Sousa JJ. A quality by design (QbD) approach in pharmaceutical development of lipid-based nanosystems: A systematic review. Journal of Drug Delivery Science and Technology. 2022 Apr 1;70:103207.
  43. Schreier H, Bouwstra J. Liposomes and niosomes as topical drug carriers: dermal and transdermal drug delivery. Journal of controlled release. 1994 Apr 1;30(1):1-5.
  44. Qumbar M, Imam SS, Ali J, Ahmad J, Ali A. Formulation and optimization of lacidipine loaded niosomal gel for transdermal delivery: in-vitro characterization and in-vivo activity. Biomedicine & Pharmacotherapy. 2017 Sep 1;93:255-66.
  45. Patel D, Patel M, Soni T, Suhagia B. Topical arginine solid lipid nanoparticles: Development and characterization by QbD approach. Journal of Drug Delivery Science and Technology. 2021 Feb 1;61:102329.
  46. Waghule T, Singhvi G, Dubey SK, Pandey MM, Gupta G, Singh M, Dua K. Microneedles: A smart approach and increasing potential for transdermal drug delivery system. Biomedicine & pharmacotherapy. 2019 Jan 1;109:1249-58.
  47. Garcês A, Amaral MH, Lobo JS, Silva AC. Formulations based on solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for cutaneous use: A review. European Journal of Pharmaceutical Sciences. 2018 Jan 15;112:159-67.
  48. Vauthier C, Bouchemal K. Methods for the preparation and manufacture of polymeric nanoparticles. Pharmaceutical research. 2009 May;26:1025-58.
  49. Mohammad IS, Hu H, Yin L, He W. Drug nanocrystals: Fabrication methods and promising therapeutic applications. International journal of pharmaceutics. 2019 May 1;562:187-202.
  50. Junghanns JU, Müller RH. Nanocrystal technology, drug delivery and clinical applications. International journal of nanomedicine. 2008 Dec 1;3(3):295-310.
  51. Gong J, Chen M, Zheng Y, Wang S, Wang Y. Polymeric micelles drug delivery system in oncology. Journal of Controlled Release. 2012 May 10;159(3):312-23.
  52. Aguilar-Jiménez Z, González-Ballesteros M, Dávila-Manzanilla SG, Espinoza-Guillén A, Ruiz-Azuara L. Development and In Vitro and In Vivo Evaluation of an Antineoplastic Copper (II) Compound (Casiopeina III-ia) Loaded in Nonionic Vesicles Using Quality by Design. International Journal of Molecular Sciences. 2022 Jan;23(21):12756.
  53. Namjoshi S, Dabbaghi M, Roberts MS, Grice JE, Mohammed Y. Quality by design: Development of the quality target product profile (QTPP) for semisolid topical products. Pharmaceutics. 2020 Mar 23;12(3):287.
  54. Poonia N, Narang JK, Lather V, Beg S, Sharma T, Singh B, Pandita D. Resveratrol loaded functionalized nanostructured lipid carriers for breast cancer targeting: Systematic development, characterization and pharmacokinetic evaluation. Colloids and Surfaces B: Biointerfaces. 2019 Sep 1;181:756-66.                    
  55. Sylvester B, Porfire A, Achim M, Rus L, Tomu?? I. A step forward towards the development of stable freeze-dried liposomes: a quality by design approach (QbD). Drug development and industrial pharmacy. 2018 Mar 4;44(3):385-97.
  56. Porfire A, Muntean DM, Rus L, Sylvester B, Tomu?? I. A quality by design approach for the development of lyophilized liposomes with simvastatin. Saudi pharmaceutical journal. 2017 Nov 1;25(7):981-92.
  57. Bragagni M, Mennini N, Furlanetto S, Orlandini S, Ghelardini C, Mura P. Development and characterization of functionalized niosomes for brain targeting of dynorphin-B. European Journal of Pharmaceutics and Biopharmaceutics. 2014 May 1;87(1):73-9.
  58. Mandpe S, Kole E, Parate V, Chatterjee A, Mujumdar A, Naik J. Design, development, and evaluation of spray dried flurbiprofen loaded sustained release polymeric nanoparticles using QBD approach to manage inflammation. Drying Technology. 2023 Nov 20;41(15):2418-30.
  59. Canchi A, Khosa A, Singhvi G, Banerjee S, Dubey SK. Design and characterization of polymeric nanoparticles of pioglitazone hydrochloride and study the effect of formulation variables using QbD approach. Current Nanomaterials. 2017 Dec 1;2(3):162-8.
  60. Vuddanda PR, Mishra A, Singh SK, Singh S. Development of polymeric nanoparticles with highly entrapped herbal hydrophilic drug using nanoprecipitation technique: an approach of quality by design. Pharmaceutical development and technology. 2015 Jul 4;20(5):579-87.
  61. Cunha S, Costa CP, Moreira JN, Lobo JM, Silva AC. Using the quality by design (QbD) approach to optimize formulations of lipid nanoparticles and nanoemulsions: A review. Nanomedicine: Nanotechnology, Biology and Medicine. 2020 Aug 1;28:102206.
  62. Vitorino C, Silva S, Gouveia F, Bicker J, Falcão A, Fortuna A. QbD-driven development of intranasal lipid nanoparticles for depression treatment. European Journal of Pharmaceutics and Biopharmaceutics. 2020 Aug 1;153:106-20.
  63. Gurumukhi VC, Bari SB. Quality by design (QbD)–based fabrication of atazanavir-loaded nanostructured lipid carriers for lymph targeting: bioavailability enhancement using chylomicron flow block model and toxicity studies. Drug Delivery and Translational Research. 2022 May;12(5):1230-52.
  64. Narayan R, Pednekar A, Bhuyan D, Gowda C, Koteshwara KB, Nayak UY. A top-down technique to improve the solubility and bioavailability of aceclofenac: in vitro and in vivo studies. International journal of nanomedicine. 2017 Jul 11:4921-35.
  65. Khurana B, Arora D, Narang RK. QbD based exploration of resveratrol loaded polymeric micelles based carbomer gel for topical treatment of plaque psoriasis: In vitro, ex vivo and in vivo studies. Journal of Drug Delivery Science and Technology. 2020 Oct 1;59:101901.
  66. Lohan S, Sharma T, Saini S, Swami R, Dhull D, Beg S, Raza K, Kumar A, Singh B. QbD-steered development of mixed nanomicelles of galantamine: Demonstration of enhanced brain uptake, prolonged systemic retention and improved biopharmaceutical attributes. International Journal of Pharmaceutics. 2021 May 1;600:120482.
  67. Sitre D, Kamble R. Articulation of quality by design elements for product development and its unique applications. Journal of Drug Delivery and Therapeutics. 2020 May 15;10(3):253-61.
  68. Shah P, Goodyear B, Haq A, Puri V, Michniak-Kohn B. Evaluations of quality by design (QbD) elements impact for developing niosomes as a promising topical drug delivery platform. Pharmaceutics. 2020 Mar 9;12(3):246.
  69. Ghose D, Patra CN, Kumar BV, Swain S, Jena BR, Choudhury P, Shree D. QbD-based formulation optimization and characterization of polymeric nanoparticles of cinacalcet hydrochloride with improved biopharmaceutical attributes. Turkish Journal of Pharmaceutical Sciences. 2021 Aug;18(4):452.
  70. Alnaim AS, Shah H, Nair AB, Mewada V, Patel S, Jacob S, Aldhubiab B, Morsy MA, Almuqbil RM, Shinu P, Shah J. Qbd-based approach to optimize niosomal gel of levosulpiride for transdermal drug delivery. Gels. 2023 Mar 10;9(3):213.
  71. Raina H, Kaur S, Jindal AB. Development of efavirenz loaded solid lipid nanoparticles: Risk assessment, quality-by-design (QbD) based optimisation and physicochemical characterisation. Journal of Drug Delivery Science and Technology. 2017 Jun 1;39:180-91.
  72. Kesharwani P, Md S, Alhakamy NA, Hosny KM, Haque A. QbD enabled azacitidine loaded liposomal nanoformulation and its in vitro evaluation. Polymers. 2021 Jan 13;13(2):250.
  73. Pallagi E, Jójárt-Laczkovich O, Németh Z, Szabó-Révész P, Csóka I. Application of the QbD-based approach in the early development of liposomes for nasal administration. International journal of pharmaceutics. 2019 May 1;562:11-22.
  74. Troiano G, Nolan J, Parsons D, Van Geen Hoven C, Zale S. A quality by design approach to developing and manufacturing polymeric nanoparticle drug products. The AAPS journal. 2016 Nov;18(6):1354-65.
  75. Gurba-Bry?kiewicz L, Maruszak W, Smuga DA, Dubiel K, Wieczorek M. Quality by Design (QbD) and Design of Experiments (DOE) as a strategy for tuning lipid nanoparticle formulations for RNA delivery. Biomedicines. 2023 Oct 11;11(10):2752.
  76. Kovács B, KOVÁCS-DEÁK BO, SZÉKELY-SZENTMIKLÓSI IS, FÜLÖP I, BÁBA LI, BODA F, PÉTERFI O. Quality-by-design in pharmaceutical development: From current perspectives to practical applications. Acta Pharmaceutica. 2021 Dec 31;71(4):497-526.
  77. Mohurle MS, Asnani MD, Chaple DR, Kurian MJ, Bais MA. Quality by Design (QbD): an emerging trend in improving quality & development of pharmaceuticals. Saudi J. Med. Pharm. Sci. 2019 Dec;5:1132-8.
  78. Kelley B, Cromwell M, Jerkins J. Integration of QbD risk assessment tools and overall risk management. Biologicals. 2016 Sep 1;44(5):341-51.
  79. Bhutani H, Kurmi M, Singh S, Beg S, Singh B. Quality by design (QbD) in analytical sciences: an overview. Quality Assurance. 2004 Sep;3:39-45.
  80. Acharya SR. Optimization of size controlled poly (lactide-co-glycolic acid) nanoparticles using quality by design concept. Asian Journal of Pharmaceutics (AJP). 2015 Jul 15:152-61.
  81. Shah B, Khunt D, Bhatt H, Misra M, Padh H. Intranasal delivery of venlafaxine loaded nanostructured lipid carrier: Risk assessment and QbD based optimization. Journal of drug delivery science and technology. 2016 Jun 1;33:37-50.
  82. Hibbert DB. Experimental design in chromatography: a tutorial review. Journal of chromatography B. 2012 Dec 1;910:2-13.
  83. Ranga S, Jaimini M, Sharma SK, Chauhan BS, Kumar A. A review on Design of Experiments (DOE). Int. J. Pharm. Chem. Sci. 2014 Jan;3(1):216-4.
  84. Badaoui FZ, Bouzid D. Formulation and Optimization of Diclofenac Sodium Loaded Ethylcellulose Nanoparticles. Brazilian Journal of Pharmaceutical Sciences. 2022 Jun 24;58:e19586.
  85. N. Politis S, Colombo P, Colombo G, M. Rekkas D. Design of experiments (DoE) in pharmaceutical development. Drug development and industrial pharmacy. 2017 Jun 3;43(6):889-901.
  86. Porter SC, Verseput RP, Cunningham CR. Process optimization using design of experiments. Pharmaceutical technology. 1997 Oct;21(10):60-71.
  87. Fukuda IM, Pinto CF, Moreira CD, Saviano AM, Lourenço FR. Design of experiments (DoE) applied to pharmaceutical and analytical quality by design (QbD). Brazilian journal of pharmaceutical sciences. 2018 Nov 8;54:e01006.
  88. Ghodake V, Vishwakarma J, Vavilala SL, Patravale V. Cefoperazone sodium liposomal formulation to mitigate P. aeruginosa biofilm in Cystic fibrosis infection: A QbD approach. International Journal of Pharmaceutics. 2020 Sep 25;587:119696.
  89. Chauhan MK, Bhatt N. Bioavailability enhancement of polymyxin B with novel drug delivery: development and optimization using quality-by-design approach. Journal of Pharmaceutical Sciences. 2019 Apr 1;108(4):1521-8.
  90. Badhwar R, Singh R, Popli H. Implementation of quality by design (qbd) approach in development of qct-smedds with combination of agnps for diabetic foot ulcer management. Indian J. Pharm. Educ. Res. 2021 Oct 1;55:1207-23.
  91. Cunha S, Costa CP, Loureiro JA, Alves J, Peixoto AF, Forbes B, Sousa Lobo JM, Silva AC. Double optimization of rivastigmine-loaded nanostructured lipid carriers (NLC) for nose-to-brain delivery using the quality by design (QbD) approach: formulation variables and instrumental parameters. Pharmaceutics. 2020 Jun 28;12(7):599.
  92. Scioli-Montoto S, Sbaraglini ML, Cisneros JS, Chain CY, Ferretti V, León IE, Alvarez VA, Castro GR, Islan GA, Talevi A, Ruiz ME. Novel phenobarbital-loaded nanostructured lipid carriers for epilepsy treatment: from QbD to in vivo evaluation. Frontiers in Chemistry. 2022 Aug 17;10:908386.
  93. Durgun ME, Mesut B, Hac?o?lu M, Güngör S, Özsoy Y. Optimization of the micellar-based in situ gelling systems posaconazole with quality by design (QbD) approach and characterization by in vitro studies. Pharmaceutics. 2022 Feb 27;14(3):526.
  94. Ohage E, Iverson R, Krummen L, Taticek R, Vega M. QbD implementation and post approval lifecycle management (PALM). Biologicals. 2016 Sep 1;44(5):332-40.
  95. Laske S, Paudel A, Scheibelhofer O, Sacher S, Hoermann T, Khinast J, Kelly A, Rantannen J, Korhonen O, Stauffer F, De Leersnyder F. A review of PAT strategies in secondary solid oral dosage manufacturing of small molecules. Journal of pharmaceutical sciences. 2017 Mar 1;106(3):667-712.
  96. Munson J, Freeman Stanfield C, Gujral B. A review of process analytical technology (PAT) in the US pharmaceutical industry. Current Pharmaceutical Analysis. 2006 Nov 1;2(4):405-14.
  97. Rathore AS, Bhambure R, Ghare V. Process analytical technology (PAT) for biopharmaceutical products. Analytical and bioanalytical chemistry. 2010 Sep;398:137-54.
  98. Scott C. Biophysical Analysis of Living Cells. Bio Process International. 2013 Jan;11:1.
  99. Bondi Jr RW, Drennen III JK. Quality by Design and the Importance of PAT in QbD. In Separation Science and Technology 2011 Jan 1 (Vol. 10, pp. 195-224). Academic Press.
  100. Besseling R, Damen M, Wijgergangs J, Hermes M, Wynia G, Gerich A. New unique PAT method and instrument for real-time inline size characterization of concentrated, flowing nanosuspensions. European Journal of Pharmaceutical Sciences. 2019 May 15;133:205-13.
  101. Schuurmans CC, Wijgergangs JP, Gerich A, Besseling R. Continuous size monitoring of turbid Titanium dioxide nanosuspensions with the Nanoflowsizer.
  102. Mészáros, L. A., Farkas, A., Madarász, L., Bicsár, R., Galata, D. L., Nagy, B., Nagy, Z. K.: UV/VIS Imaging-Based PAT Tool for Drug Particle Size Inspection in Intact Tablets Supported by Pattern Recognition Neural Networks, Int. J. Pharm., 2022, 620 121773.
  103. Gerzon G, Sheng Y, Kirkitadze M. Process analytical technologies–advances in bioprocess integration and future perspectives. Journal of pharmaceutical and biomedical analysis. 2022 Jan 5;207:114379.

Reference

  1. Yu LX, Amidon G, Khan MA, Hoag SW, Polli J, Raju GK, Woodcock J. Understanding pharmaceutical quality by design. The AAPS journal. 2014 Jul;16:771-83.
  2. Zhang L, Mao S. Application of quality by design in the current drug development. Asian journal of pharmaceutical sciences. 2017 Jan 1;12(1):1-8.
  3. Thomson DS, Austin SA, Devine-Wright H, Mills GR. Managing value and quality in design. Building Research & Information. 2003 Jan 1;31(5):334-45.
  4. Beg S, Rahman M, Kohli K. Quality-by-design approach as a systematic tool for the development of nanopharmaceutical products. Drug discovery today. 2019 Mar 1;24(3):717-25.
  5. Sangshetti JN, Deshpande M, Zaheer Z, Shinde DB, Arote R. Quality by design approach: Regulatory need. Arabian Journal of chemistry. 2017 May 1;10:S3412-25.
  6. Bhoop BS. Quality by Design (QbD) for holistic pharma excellence and regulatory compliance. Pharm times. 2014;46(8):26-33.
  7. Juran JM. Juran on quality by design: The new steps for planning quality into goods and services. Simon & Schuster; 1992.
  8. Bala I, Verma S, Namdev KK. A Review on Process Variables Effects of an Oral Dosage Form by Using “Quality by Design”. International Journal of Drug Delivery Technology. 2015;5(1).
  9. Lionberger RA, Lee SL, Lee L, Raw A, Yu LX. Quality by design: concepts for ANDAs. The AAPS journal. 2008 Jun;10:268-76.
  10. Jeevanandam J, San Chan Y, Danquah MK. Nano-formulations of drugs: recent developments, impact and challenges. Biochimie. 2016 Sep 1;128:99-112.
  11. Rathore AS, Winkle H. Quality by design for biopharmaceuticals. Nature biotechnology. 2009 Jan;27(1):26-34.
  12. Borman P, Chatfield M, Nethercote P, Thompson D, Truman K. The application of quality by design to analytical methods. Pharmaceutical Technology. 2007 Oct 1;31(10):142-8.
  13. Javed MN, Alam MS, Waziri A, Pottoo FH, Yadav AK, Hasnain MS, Almalki FA. QbD applications for the development of nanopharmaceutical products. InPharmaceutical quality by design 2019 Jan 1 (pp. 229-253). Academic Press.
  14. Tefas LR, RUS L, Achim M, Vlase L, TOMU?? I. APPLICATION OF THE QUALITY BY DESIGN CONCEPT IN THE DEVELOPMENT OF QUERCETIN-LOADED POLYMERIC NANOPARTICLES. Farmacia. 2018 Sep 1;66(5).
  15. Patil AS, Pethe AM. Quality by Design (QbD): A new concept for development of quality pharmaceuticals. International journal of pharmaceutical quality assurance. 2013 Apr;4(2):13-9.
  16. Yu LX. Pharmaceutical quality by design: product and process development, understanding, and control. Pharmaceutical research. 2008 Apr;25:781-91.
  17. Nadpara NP, Thumar RV, Kalola VN, Patel PB. Quality by design (QBD): A complete review. Int J Pharm Sci Rev Res. 2012;17(2):20-8.
  18. ICH Harmonised Tripartite Guideline Q8 (R2) on Pharmaceutical Development. Available at: https://database.ich.org/sites/default/files/Q8_R2_Guideline.pdf. Accessed January 2024.
  19. ICH Harmonised Tripartite Guideline Q9 on Quality Risk Management. Available at: https://database.ich.org/sites/default/files/Q9_Guideline.pdf. Accessed January 2024.
  20. Reddy VV, Vishal Gupta N, Raghunandan HV, Nitin Kashyap U. Quality risk management in pharmaceutical industry: a review. International Journal of Pharm Tech Research. 2014;6(3):908-14
  21. Hasnain MS, Siddiqui S, Rao S, Mohanty P, Jahan Ara T, Beg S. QbD-driven development and validation of a bioanalytical LC–MS method for quantification of fluoxetine in human plasma. Journal of Chromatographic Science. 2016 May 1;54(5):736-43.
  22. Short SM, Cogdill RP, DRENNEN III JK, ANDERSON CA. A New Definition of Pharmaceutical Quality: Assembly of a Risk Simulation Platform to Investigate the Impact of Manufacturing/Product Variabcpility on Clinical Performance. Journal of pharmaceutical sciences. 2010 Dec 1;99(12):5046-59.
  23. Haleem RM, Salem MY, Fatahallah FA, Abdelfattah LE. Quality in the pharmaceutical industry–A literature review. Saudi pharmaceutical journal. 2015 Oct 1;23(5):463-9.
  24. Madaan T, Pandey S, Talegaonkar S, Delhi N. Nanotechnology: A smart drug delivery tool in modern healthcare. Journal of Chemical and Pharmaceutical Research. 2015;7(6):257-64.
  25. Khiev D, Mohamed ZA, Vichare R, Paulson R, Bhatia S, Mohapatra S, Lobo GP, Valapala M, Kerur N, Passaglia CL, Mohapatra SS. Emerging nano-formulations and nanomedicines applications for ocular drug delivery. Nanomaterials. 2021 Jan 12;11(1):173.
  26. Zhou X, Hao Y, Yuan L, Pradhan S, Shrestha K, Pradhan O, Liu H, Li W. Nano-formulations for transdermal drug delivery: A review. Chinese Chemical Letters. 2018 Dec 1;29(12):1713-24.
  27. Pathak K, Raghuvanshi S. Oral bioavailability: issues and solutions via nanoformulations. Clinical pharmacokinetics. 2015 Apr;54:325-57.
  28. Jiang W, Kim S, Zhang X, Lionberger RA, Davit BM, Conner DP, Lawrence XY. The role of predictive biopharmaceutical modeling and simulation in drug development and regulatory evaluation. International journal of pharmaceutics. 2011 Oct 14;418(2):151-60.
  29. Farjadian F, Ghasemi A, Gohari O, Roointan A, Karimi M, Hamblin MR. Nanopharmaceuticals and nanomedicines currently on the market: challenges and opportunities. Nanomedicine. 2019 Jan 1;14(1):93-126.
  30. Patra JK, Das G, Fraceto LF, Campos EV, Rodriguez-Torres MD, Acosta-Torres LS, Diaz-Torres LA, Grillo R, Swamy MK, Sharma S, Habtemariam S. Nano based drug delivery systems: recent developments and future prospects. Journal of nanobiotechnology. 2018 Dec;16:1-33.
  31. Peltonen L. Design space and QbD approach for production of drug nanocrystals by wet media milling techniques. Pharmaceutics. 2018 Jul 25;10(3):104.
  32. Ragelle H, Danhier F, Préat V, Langer R, Anderson DG. Nanoparticle-based drug delivery systems: a commercial and regulatory outlook as the field matures. Expert opinion on drug delivery. 2017 Jul 3;14(7):851-64.                 
  33. Chavda VP. Nanobased nano drug delivery: a comprehensive review. Applications of targeted nano drugs and delivery systems. 2019 Jan 1:69-92.
  34. Birla D, Khandale N, Bashir B, ShahbazAlam M, Vishwas S, Gupta G, Dureja H, Kumbhar PS, Disouza J, Patravale V, Veiga F. Application of quality by design in optimization of nanoformulations: Principle, perspectives and practices. Drug Delivery and Translational Research. 2024 Aug 10:1-33.
  35. Zieli?ska A, Carreiró F, Oliveira AM, Neves A, Pires B, Venkatesh DN, Durazzo A, Lucarini M, Eder P, Silva AM, Santini A. Polymeric nanoparticles: production, characterization, toxicology and ecotoxicology. Molecules. 2020 Aug 15;25(16):3731.
  36. Singh B, Sharma T, Saini S, Kaur R, Jain A, Raza K, Beg S. Systematic development of drug nanocargos using formulation by design (FbD): an updated overview. Critical Reviews™ in Therapeutic Drug Carrier Systems. 2020;37(3).
  37. Sylvester B, Porfire A, Muntean DM, Vlase L, Lupu? L, Licarete E, Sesarman A, Alupei MC, Banciu M, Achim M, Tomu?? I. Optimization of prednisolone-loaded long-circulating liposomes via application of Quality by Design (QbD) approach. Journal of liposome research. 2018 Jan 2;28(1):49-61.
  38. Simoes A, Veiga F, Figueiras A, Vitorino C. A practical framework for implementing Quality by Design to the development of topical drug products: Nanosystem-based dosage forms. International journal of pharmaceutics. 2018 Sep 5;548(1):385-99.
  39. Dobó DG, Németh Z, Sipos B, Cseh M, Pallagi E, Berkesi D, Kozma G, Kónya Z, Csóka I. Pharmaceutical development and design of thermosensitive liposomes based on the QbD approach. Molecules. 2022 Feb 24;27(5):1536.
  40. Charoo NA, Shamsher AA, Zidan AS, Rahman Z. Quality by design approach for formulation development: a case study of dispersible tablets. International journal of pharmaceutics. 2012 Feb 28;423(2):167-78.
  41. Bartelds R, Nematollahi MH, Pols T, Stuart MC, Pardakhty A, Asadikaram G, Poolman B. Niosomes, an alternative for liposomal delivery. PLoS One. 2018 Apr 12;13(4):e0194179.
  42. Zagalo DM, Silva BM, Silva C, Simoes S, Sousa JJ. A quality by design (QbD) approach in pharmaceutical development of lipid-based nanosystems: A systematic review. Journal of Drug Delivery Science and Technology. 2022 Apr 1;70:103207.
  43. Schreier H, Bouwstra J. Liposomes and niosomes as topical drug carriers: dermal and transdermal drug delivery. Journal of controlled release. 1994 Apr 1;30(1):1-5.
  44. Qumbar M, Imam SS, Ali J, Ahmad J, Ali A. Formulation and optimization of lacidipine loaded niosomal gel for transdermal delivery: in-vitro characterization and in-vivo activity. Biomedicine & Pharmacotherapy. 2017 Sep 1;93:255-66.
  45. Patel D, Patel M, Soni T, Suhagia B. Topical arginine solid lipid nanoparticles: Development and characterization by QbD approach. Journal of Drug Delivery Science and Technology. 2021 Feb 1;61:102329.
  46. Waghule T, Singhvi G, Dubey SK, Pandey MM, Gupta G, Singh M, Dua K. Microneedles: A smart approach and increasing potential for transdermal drug delivery system. Biomedicine & pharmacotherapy. 2019 Jan 1;109:1249-58.
  47. Garcês A, Amaral MH, Lobo JS, Silva AC. Formulations based on solid lipid nanoparticles (SLN) and nanostructured lipid carriers (NLC) for cutaneous use: A review. European Journal of Pharmaceutical Sciences. 2018 Jan 15;112:159-67.
  48. Vauthier C, Bouchemal K. Methods for the preparation and manufacture of polymeric nanoparticles. Pharmaceutical research. 2009 May;26:1025-58.
  49. Mohammad IS, Hu H, Yin L, He W. Drug nanocrystals: Fabrication methods and promising therapeutic applications. International journal of pharmaceutics. 2019 May 1;562:187-202.
  50. Junghanns JU, Müller RH. Nanocrystal technology, drug delivery and clinical applications. International journal of nanomedicine. 2008 Dec 1;3(3):295-310.
  51. Gong J, Chen M, Zheng Y, Wang S, Wang Y. Polymeric micelles drug delivery system in oncology. Journal of Controlled Release. 2012 May 10;159(3):312-23.
  52. Aguilar-Jiménez Z, González-Ballesteros M, Dávila-Manzanilla SG, Espinoza-Guillén A, Ruiz-Azuara L. Development and In Vitro and In Vivo Evaluation of an Antineoplastic Copper (II) Compound (Casiopeina III-ia) Loaded in Nonionic Vesicles Using Quality by Design. International Journal of Molecular Sciences. 2022 Jan;23(21):12756.
  53. Namjoshi S, Dabbaghi M, Roberts MS, Grice JE, Mohammed Y. Quality by design: Development of the quality target product profile (QTPP) for semisolid topical products. Pharmaceutics. 2020 Mar 23;12(3):287.
  54. Poonia N, Narang JK, Lather V, Beg S, Sharma T, Singh B, Pandita D. Resveratrol loaded functionalized nanostructured lipid carriers for breast cancer targeting: Systematic development, characterization and pharmacokinetic evaluation. Colloids and Surfaces B: Biointerfaces. 2019 Sep 1;181:756-66.                    
  55. Sylvester B, Porfire A, Achim M, Rus L, Tomu?? I. A step forward towards the development of stable freeze-dried liposomes: a quality by design approach (QbD). Drug development and industrial pharmacy. 2018 Mar 4;44(3):385-97.
  56. Porfire A, Muntean DM, Rus L, Sylvester B, Tomu?? I. A quality by design approach for the development of lyophilized liposomes with simvastatin. Saudi pharmaceutical journal. 2017 Nov 1;25(7):981-92.
  57. Bragagni M, Mennini N, Furlanetto S, Orlandini S, Ghelardini C, Mura P. Development and characterization of functionalized niosomes for brain targeting of dynorphin-B. European Journal of Pharmaceutics and Biopharmaceutics. 2014 May 1;87(1):73-9.
  58. Mandpe S, Kole E, Parate V, Chatterjee A, Mujumdar A, Naik J. Design, development, and evaluation of spray dried flurbiprofen loaded sustained release polymeric nanoparticles using QBD approach to manage inflammation. Drying Technology. 2023 Nov 20;41(15):2418-30.
  59. Canchi A, Khosa A, Singhvi G, Banerjee S, Dubey SK. Design and characterization of polymeric nanoparticles of pioglitazone hydrochloride and study the effect of formulation variables using QbD approach. Current Nanomaterials. 2017 Dec 1;2(3):162-8.
  60. Vuddanda PR, Mishra A, Singh SK, Singh S. Development of polymeric nanoparticles with highly entrapped herbal hydrophilic drug using nanoprecipitation technique: an approach of quality by design. Pharmaceutical development and technology. 2015 Jul 4;20(5):579-87.
  61. Cunha S, Costa CP, Moreira JN, Lobo JM, Silva AC. Using the quality by design (QbD) approach to optimize formulations of lipid nanoparticles and nanoemulsions: A review. Nanomedicine: Nanotechnology, Biology and Medicine. 2020 Aug 1;28:102206.
  62. Vitorino C, Silva S, Gouveia F, Bicker J, Falcão A, Fortuna A. QbD-driven development of intranasal lipid nanoparticles for depression treatment. European Journal of Pharmaceutics and Biopharmaceutics. 2020 Aug 1;153:106-20.
  63. Gurumukhi VC, Bari SB. Quality by design (QbD)–based fabrication of atazanavir-loaded nanostructured lipid carriers for lymph targeting: bioavailability enhancement using chylomicron flow block model and toxicity studies. Drug Delivery and Translational Research. 2022 May;12(5):1230-52.
  64. Narayan R, Pednekar A, Bhuyan D, Gowda C, Koteshwara KB, Nayak UY. A top-down technique to improve the solubility and bioavailability of aceclofenac: in vitro and in vivo studies. International journal of nanomedicine. 2017 Jul 11:4921-35.
  65. Khurana B, Arora D, Narang RK. QbD based exploration of resveratrol loaded polymeric micelles based carbomer gel for topical treatment of plaque psoriasis: In vitro, ex vivo and in vivo studies. Journal of Drug Delivery Science and Technology. 2020 Oct 1;59:101901.
  66. Lohan S, Sharma T, Saini S, Swami R, Dhull D, Beg S, Raza K, Kumar A, Singh B. QbD-steered development of mixed nanomicelles of galantamine: Demonstration of enhanced brain uptake, prolonged systemic retention and improved biopharmaceutical attributes. International Journal of Pharmaceutics. 2021 May 1;600:120482.
  67. Sitre D, Kamble R. Articulation of quality by design elements for product development and its unique applications. Journal of Drug Delivery and Therapeutics. 2020 May 15;10(3):253-61.
  68. Shah P, Goodyear B, Haq A, Puri V, Michniak-Kohn B. Evaluations of quality by design (QbD) elements impact for developing niosomes as a promising topical drug delivery platform. Pharmaceutics. 2020 Mar 9;12(3):246.
  69. Ghose D, Patra CN, Kumar BV, Swain S, Jena BR, Choudhury P, Shree D. QbD-based formulation optimization and characterization of polymeric nanoparticles of cinacalcet hydrochloride with improved biopharmaceutical attributes. Turkish Journal of Pharmaceutical Sciences. 2021 Aug;18(4):452.
  70. Alnaim AS, Shah H, Nair AB, Mewada V, Patel S, Jacob S, Aldhubiab B, Morsy MA, Almuqbil RM, Shinu P, Shah J. Qbd-based approach to optimize niosomal gel of levosulpiride for transdermal drug delivery. Gels. 2023 Mar 10;9(3):213.
  71. Raina H, Kaur S, Jindal AB. Development of efavirenz loaded solid lipid nanoparticles: Risk assessment, quality-by-design (QbD) based optimisation and physicochemical characterisation. Journal of Drug Delivery Science and Technology. 2017 Jun 1;39:180-91.
  72. Kesharwani P, Md S, Alhakamy NA, Hosny KM, Haque A. QbD enabled azacitidine loaded liposomal nanoformulation and its in vitro evaluation. Polymers. 2021 Jan 13;13(2):250.
  73. Pallagi E, Jójárt-Laczkovich O, Németh Z, Szabó-Révész P, Csóka I. Application of the QbD-based approach in the early development of liposomes for nasal administration. International journal of pharmaceutics. 2019 May 1;562:11-22.
  74. Troiano G, Nolan J, Parsons D, Van Geen Hoven C, Zale S. A quality by design approach to developing and manufacturing polymeric nanoparticle drug products. The AAPS journal. 2016 Nov;18(6):1354-65.
  75. Gurba-Bry?kiewicz L, Maruszak W, Smuga DA, Dubiel K, Wieczorek M. Quality by Design (QbD) and Design of Experiments (DOE) as a strategy for tuning lipid nanoparticle formulations for RNA delivery. Biomedicines. 2023 Oct 11;11(10):2752.
  76. Kovács B, KOVÁCS-DEÁK BO, SZÉKELY-SZENTMIKLÓSI IS, FÜLÖP I, BÁBA LI, BODA F, PÉTERFI O. Quality-by-design in pharmaceutical development: From current perspectives to practical applications. Acta Pharmaceutica. 2021 Dec 31;71(4):497-526.
  77. Mohurle MS, Asnani MD, Chaple DR, Kurian MJ, Bais MA. Quality by Design (QbD): an emerging trend in improving quality & development of pharmaceuticals. Saudi J. Med. Pharm. Sci. 2019 Dec;5:1132-8.
  78. Kelley B, Cromwell M, Jerkins J. Integration of QbD risk assessment tools and overall risk management. Biologicals. 2016 Sep 1;44(5):341-51.
  79. Bhutani H, Kurmi M, Singh S, Beg S, Singh B. Quality by design (QbD) in analytical sciences: an overview. Quality Assurance. 2004 Sep;3:39-45.
  80. Acharya SR. Optimization of size controlled poly (lactide-co-glycolic acid) nanoparticles using quality by design concept. Asian Journal of Pharmaceutics (AJP). 2015 Jul 15:152-61.
  81. Shah B, Khunt D, Bhatt H, Misra M, Padh H. Intranasal delivery of venlafaxine loaded nanostructured lipid carrier: Risk assessment and QbD based optimization. Journal of drug delivery science and technology. 2016 Jun 1;33:37-50.
  82. Hibbert DB. Experimental design in chromatography: a tutorial review. Journal of chromatography B. 2012 Dec 1;910:2-13.
  83. Ranga S, Jaimini M, Sharma SK, Chauhan BS, Kumar A. A review on Design of Experiments (DOE). Int. J. Pharm. Chem. Sci. 2014 Jan;3(1):216-4.
  84. Badaoui FZ, Bouzid D. Formulation and Optimization of Diclofenac Sodium Loaded Ethylcellulose Nanoparticles. Brazilian Journal of Pharmaceutical Sciences. 2022 Jun 24;58:e19586.
  85. N. Politis S, Colombo P, Colombo G, M. Rekkas D. Design of experiments (DoE) in pharmaceutical development. Drug development and industrial pharmacy. 2017 Jun 3;43(6):889-901.
  86. Porter SC, Verseput RP, Cunningham CR. Process optimization using design of experiments. Pharmaceutical technology. 1997 Oct;21(10):60-71.
  87. Fukuda IM, Pinto CF, Moreira CD, Saviano AM, Lourenço FR. Design of experiments (DoE) applied to pharmaceutical and analytical quality by design (QbD). Brazilian journal of pharmaceutical sciences. 2018 Nov 8;54:e01006.
  88. Ghodake V, Vishwakarma J, Vavilala SL, Patravale V. Cefoperazone sodium liposomal formulation to mitigate P. aeruginosa biofilm in Cystic fibrosis infection: A QbD approach. International Journal of Pharmaceutics. 2020 Sep 25;587:119696.
  89. Chauhan MK, Bhatt N. Bioavailability enhancement of polymyxin B with novel drug delivery: development and optimization using quality-by-design approach. Journal of Pharmaceutical Sciences. 2019 Apr 1;108(4):1521-8.
  90. Badhwar R, Singh R, Popli H. Implementation of quality by design (qbd) approach in development of qct-smedds with combination of agnps for diabetic foot ulcer management. Indian J. Pharm. Educ. Res. 2021 Oct 1;55:1207-23.
  91. Cunha S, Costa CP, Loureiro JA, Alves J, Peixoto AF, Forbes B, Sousa Lobo JM, Silva AC. Double optimization of rivastigmine-loaded nanostructured lipid carriers (NLC) for nose-to-brain delivery using the quality by design (QbD) approach: formulation variables and instrumental parameters. Pharmaceutics. 2020 Jun 28;12(7):599.
  92. Scioli-Montoto S, Sbaraglini ML, Cisneros JS, Chain CY, Ferretti V, León IE, Alvarez VA, Castro GR, Islan GA, Talevi A, Ruiz ME. Novel phenobarbital-loaded nanostructured lipid carriers for epilepsy treatment: from QbD to in vivo evaluation. Frontiers in Chemistry. 2022 Aug 17;10:908386.
  93. Durgun ME, Mesut B, Hac?o?lu M, Güngör S, Özsoy Y. Optimization of the micellar-based in situ gelling systems posaconazole with quality by design (QbD) approach and characterization by in vitro studies. Pharmaceutics. 2022 Feb 27;14(3):526.
  94. Ohage E, Iverson R, Krummen L, Taticek R, Vega M. QbD implementation and post approval lifecycle management (PALM). Biologicals. 2016 Sep 1;44(5):332-40.
  95. Laske S, Paudel A, Scheibelhofer O, Sacher S, Hoermann T, Khinast J, Kelly A, Rantannen J, Korhonen O, Stauffer F, De Leersnyder F. A review of PAT strategies in secondary solid oral dosage manufacturing of small molecules. Journal of pharmaceutical sciences. 2017 Mar 1;106(3):667-712.
  96. Munson J, Freeman Stanfield C, Gujral B. A review of process analytical technology (PAT) in the US pharmaceutical industry. Current Pharmaceutical Analysis. 2006 Nov 1;2(4):405-14.
  97. Rathore AS, Bhambure R, Ghare V. Process analytical technology (PAT) for biopharmaceutical products. Analytical and bioanalytical chemistry. 2010 Sep;398:137-54.
  98. Scott C. Biophysical Analysis of Living Cells. Bio Process International. 2013 Jan;11:1.
  99. Bondi Jr RW, Drennen III JK. Quality by Design and the Importance of PAT in QbD. In Separation Science and Technology 2011 Jan 1 (Vol. 10, pp. 195-224). Academic Press.
  100. Besseling R, Damen M, Wijgergangs J, Hermes M, Wynia G, Gerich A. New unique PAT method and instrument for real-time inline size characterization of concentrated, flowing nanosuspensions. European Journal of Pharmaceutical Sciences. 2019 May 15;133:205-13.
  101. Schuurmans CC, Wijgergangs JP, Gerich A, Besseling R. Continuous size monitoring of turbid Titanium dioxide nanosuspensions with the Nanoflowsizer.
  102. Mészáros, L. A., Farkas, A., Madarász, L., Bicsár, R., Galata, D. L., Nagy, B., Nagy, Z. K.: UV/VIS Imaging-Based PAT Tool for Drug Particle Size Inspection in Intact Tablets Supported by Pattern Recognition Neural Networks, Int. J. Pharm., 2022, 620 121773.
  103. Gerzon G, Sheng Y, Kirkitadze M. Process analytical technologies–advances in bioprocess integration and future perspectives. Journal of pharmaceutical and biomedical analysis. 2022 Jan 5;207:114379.

Photo
M. M. Chogale
Corresponding author

Department of Pharmaceutics, Saraswathi Vidya Bhawan’s College of Pharmacy, Sankara Nagar, Dombivali (East) - 421204, Dist: Thane, State: Maharashtra, India

Photo
B. M. N. Shaikh
Co-author

Department of Pharmaceutics, Saraswathi Vidya Bhawan’s College of Pharmacy, Sankara Nagar, Dombivali (East) - 421204, Dist: Thane, State: Maharashtra, India

Photo
A. R. Gupta
Co-author

Department of Pharmaceutics, Saraswathi Vidya Bhawan’s College of Pharmacy, Sankara Nagar, Dombivali (East) - 421204, Dist: Thane, State: Maharashtra, India

Photo
S. P. Udeg
Co-author

Department of Pharmaceutics, Saraswathi Vidya Bhawan’s College of Pharmacy, Sankara Nagar, Dombivali (East) - 421204, Dist: Thane, State: Maharashtra, India

Photo
V. B. Patil
Co-author

Department of Pharmaceutics, Saraswathi Vidya Bhawan’s College of Pharmacy, Sankara Nagar, Dombivali (East) - 421204, Dist: Thane, State: Maharashtra, India

Photo
A. S. Jagtap
Co-author

Department of Pharmaceutics, Saraswathi Vidya Bhawan’s College of Pharmacy, Sankara Nagar, Dombivali (East) - 421204, Dist: Thane, State: Maharashtra, India

M. M. Chogale, B. M. N. Shaikh, A. R. Gupta, S. P. Udeg, V. B. Patil, A. S. Jagtap, A Comprehensive QBD Strategy for Nanotherapeutics Development: A Review, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 01, 315-333. https://doi.org/10.5281/zenodo.14605197

More related articles
Development of a rapid LC method for Metopimazine ...
Dr.Mukesh S.Patil , Sonal B. Bangar , Tanmay Kamble , Dr.Ashish J...
Advancements and Applications of Transdermal Drug ...
Sharvari Gurav , Shruti Kalebere , Sudesh Patil, Umesh Kolap , Dr...
The Pathogenesis, Diagnosis, Treatment and Future ...
Sabafarin Shaikh, Prachi Akolkar, Manisha Ravate, Monika Gaikwad,...
Review On Transdermal Drug Delivery System...
Sachin Pawar, Nitin Sawle, Dr. Gajanan Sanap, ...
Recent Advancement: 3D-Printed Orodispersible Film as Pharmaceutical Dosage Form...
Bhavesh Machhi , Aathira Chandran, Divyanka Bodas, Chandrakant Wadile, Pankaj Mandpe, ...
Pharmacogenomic Testing in a Bangladeshi Patient with Recurrent Cardiovascular E...
Marjia Islam Jemi, Shamim Ahmed, Tariqul Islam , Nur A. Samira , ...
Related Articles
A Comprehensive Review On Quality By Design (QbD)...
Nagineni Sudarshan Rao, Katta Subramanya Swamy, ...
Formulation And Evaluations Of Beet Root Herbal Lipstick...
Gayatri D. karwate , Prajakta V. Muthal , Swati P. Deshmukh, Madhuri B. Wankhade, ...
A Review On Recent Advances In Development Of RP-HPLC Method...
Madhuri R. Shirsath , Sonali A. Waghmare, Pradyumna P. Ige, ...
Formulation And Evaluation Study Of Herbal Buccal Patch For Mouth Ulcer With Cur...
Daphne Sherine S, Ravichandran S., Abjel A., Gopi S., Sukesh Kumar B., ...
Development of a rapid LC method for Metopimazine based on a Quality by Design (...
Dr.Mukesh S.Patil , Sonal B. Bangar , Tanmay Kamble , Dr.Ashish Jain, ...
More related articles
Development of a rapid LC method for Metopimazine based on a Quality by Design (...
Dr.Mukesh S.Patil , Sonal B. Bangar , Tanmay Kamble , Dr.Ashish Jain, ...
Advancements and Applications of Transdermal Drug Delivery Systems (TDDS) ...
Sharvari Gurav , Shruti Kalebere , Sudesh Patil, Umesh Kolap , Dr. Shobhraj Malavi , ...
The Pathogenesis, Diagnosis, Treatment and Future Prospectives in Pharmaceutical...
Sabafarin Shaikh, Prachi Akolkar, Manisha Ravate, Monika Gaikwad, Rohini A. Satdive, Renuka K. Sagne...
Development of a rapid LC method for Metopimazine based on a Quality by Design (...
Dr.Mukesh S.Patil , Sonal B. Bangar , Tanmay Kamble , Dr.Ashish Jain, ...
Advancements and Applications of Transdermal Drug Delivery Systems (TDDS) ...
Sharvari Gurav , Shruti Kalebere , Sudesh Patil, Umesh Kolap , Dr. Shobhraj Malavi , ...
The Pathogenesis, Diagnosis, Treatment and Future Prospectives in Pharmaceutical...
Sabafarin Shaikh, Prachi Akolkar, Manisha Ravate, Monika Gaikwad, Rohini A. Satdive, Renuka K. Sagne...