Sunteți pe pagina 1din 10

REVIEW

Excipient Variability and Its Impact on Dosage Form Functionality


VIVEK S. DAVE,1 SUPRIT D. SAOJI,2 NISHIKANT A. RAUT,2 RAHUL V. HAWARE3
1
St. John Fisher College, Wegmans School of Pharmacy, Rochester, New York, USA
2
Department of Pharmaceutical Sciences, R.T.M. Nagpur University, Nagpur, India
3
Campbell University College of Pharmacy and Health Science, Buies Creek, North Carolina, USA

Received 15 September 2014; revised 30 October 2014; accepted 14 November 2014


Published online in Wiley Online Library (wileyonlinelibrary.com). DOI 10.1002/jps.24299

ABSTRACT: Pharmaceutical excipients are essential components of most modern dosage forms. Although defined as pharmacologically
inert, excipients can be thought of as the true enablers of drug product performance. Unintentional variability in the properties of the
excipients may be unavoidable, albeit minimizable. The variability may originate from the source, the excipient-manufacturing process, or
during the manufacturing of dosage forms. Excipient variability may have a range of influences on their functionality and performance in the
dosage form. A better understanding of these influences on the critical quality attributes of the final product is of prime importance. Modern
analytical tools provide a significant assistance in characterizing excipient variability to achieve this understanding. The principles and
concepts of Quality-by-Design, process analytical technology, and design space, provide a holistic risk-based approach toward manufacture
and application of excipients in pharmaceutical formulations. The International Pharmaceutical Excipients Council (IPEC) has developed
guidelines for proper selection, use, and evaluation of excipients in pharmaceutical products.  C 2015 Wiley Periodicals, Inc. and the

American Pharmacists Association J Pharm Sci


Keywords: excipients; formulation; physical characterization; physicochemical properties; process analytical technology (PAT); Quality
by design (QBD)

INTRODUCTION from various sources, including plant, animal, mineral, biotech-


nology, and chemical synthesis. Pharmaceutical excipients are
Various agencies like the United States Food and Drug Ad-
a diverse group of materials such as solids, semisolids, liquids,
ministration (US FDA), the United States Pharmacopeia
and gases. Each excipient has a unique development process
(USP), and the International Pharmaceutical Excipient Council
and associated know-how. Excipients are used in a variety of
(IPEC) broadly define pharmaceutical excipients as any phar-
dosage forms and routes of administration. Excipient man-
macologically inert component other than the active pharma-
ufacturing processes typically involve harvesting, extraction,
ceutical ingredient (API) added intentionally to the pharma-
synthetic chemistry, agglomeration, size reduction, and fer-
ceutical formulations.1 These intentions are associated with
mentation. They are quite often manufactured on dedicated
one or more domains of the drug product development. Excip-
equipment, frequently using some form of continuous or batch
ients are used to improve processing (e.g., improving powder
manufacturing. The scale of manufacture may be very different
flow,2,3 powder compactibility,4–6 etc.), enhance aesthetics (e.g.,
to that typically encountered in the manufacturing of a phar-
identification,7 branding, etc.), optimize product performance
maceutical drug product.16 The discovery and development of
(e.g., modified drug release8–11 ), and/or to facilitate patient com-
a new chemical excipient undergoes an extensive regulatory
pliance (e.g., taste masking12–15 ). Excipients are found ubiqui-
review process similar to a new drug.17 This is carried out to
tously as an indispensable part of virtually every pharmaceu-
evaluate safety and toxicity in humans. Thus, all excipients are
tical formulation to serve one or more of the above-mentioned
required to fulfill a plethora of safety, quality, and functionality
purposes. They may constitute anywhere from 1% to 99% of
criteria issued by regulatory agencies before their adoption in
the total formulation mass. Therefore, excipient properties may
actual formulations. The US FDA maintains a database of ex-
have significant impact on the resulting formulation properties.
cipients (inactive ingredients or IIG) present in approved drug
As a major component of the pharmaceutical development,
products. This database serves as a guideline to the preformu-
preformulation scientists invest a substantial amount of time
lation and formulation scientists for a rational selection and
on the careful evaluation of various excipient-related parame-
use of excipients in the drug product. The approval of an ex-
ters such as drug–excipient compatibility and excipient pro-
cipient by the FDA for implementation in formulation design
cessability. Furthermore, the cost of an excipient and its
entails cross-referencing to pharma/food/cosmetic compendia.
consistent availability has been other important factor in the
Additionally, excipients are cross-referenced in an abbreviated
excipient selection.
new drug application, new drug application, and/or biologic li-
A majority of pharmaceutical excipients are supplied by the
cense application for a particular function in a drug product.
food and the chemical industry. Excipients are manufactured
Excipient approval also includes announcing of substances to
be “GRAS” (generally recognized as safe) or approving a “FAP”
(food additive petition).18
Correspondence to: Vivek S. Dave (Telephone: +443-789-8649; Fax: +585-385-
5295; E-mail: viveksdave@gmail.com)
The commercial manufacture of excipients is required to fol-
Journal of Pharmaceutical Sciences
low stringent current good manufacturing practices (cGMP)

C 2015 Wiley Periodicals, Inc. and the American Pharmacists Association guidelines and meet specifications regarding residuals (e.g.,

Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES 1


2 REVIEW

Figure 1. The impact of excipient variability on the CQAs of the drug product.

ethylene glycol in glycerine), degradation products (e.g., 5- API and impact of the process parameters on the API is well
hydroxymethyl furfural in lactose), and so on. They require a understood and controlled. Excipients comprise a substantial
strict control over lot-to-lot excipient variability associated with mass of the formulation. As shown in Figure 1, excipients may
inherent discrepancies in the excipient sources. Manufacturers have a significant contribution to the variability in the criti-
need to implement built-in traceability in the supply chain to cal quality attributes (CQAs) of the final drug product. Thus,
ensure control of the overall quality. This is necessary to main- it is equally important to understand the impact of excipient
tain safety, quality, and consistency in the product. Recently, it variability on the drug product manufacturability and perfor-
has become increasingly important to clearly define the func- mance.
tion of an excipient in a formulation along with the knowledge A sound understanding of the raw material and process vari-
of factors influencing its functionality. These requirements, to abilities is essential to define a design space for a robust for-
a large extent, avoid unnecessary excipient-related variations mulation development. This can be achieved with two common
in the formulations apart from the APIs. approaches.22 The traditional approach involves narrowing the
Other challenges include cost and awareness. Newer excip- specifications with a tight control of input parameters, includ-
ients typically tend to be more expensive than traditionally ing the API, the excipients, and the process variables. This
used excipients because of the associated development costs. approach may not necessarily address the interactions of the
The excipient database needs to be updated often, a lack of influencing variables. The sum of interactions, called interac-
which may result in formulators not being aware of newly de- tion factor, may lead to potential problems in developing a ro-
veloped excipients with improved functionalities. Additionally, bust formulation. Another more modern approach is based on a
limited manufacturing history [“Quality-by-Design” (QbD) type feed-forward control. In this approach, the assumed input vari-
studies] of new excipients presents an inherent risk of sourcing ability is incorporated into the design space. This is followed by
and variability to the formulators. Thus, there exist develop- utilizing a sufficient understanding of the process to define an
ment and regulatory risks that may act as barriers to the use appropriate process end point. The second approach appears
of excipients, particularly newer excipients. This review aims to match the QbD intent in a better way. This approach might
to provide a comprehensive overview of the trends in excipient provide a larger design space with a more flexible formulation
use and the influence of excipient variability on dosage form and process.22–24
functionality. Quality-by-Design is defined as a scientific, risk-based, holis-
Pharmaceutical drug products are expected to deliver the tic, and proactive approach to pharmaceutical development.25
drugs to the patient in a required amount, required rate, and QbD along with process analytical technology (PAT) tools aims
with the same potency throughout their shelf-life. This depends to achieve a thorough understanding of the CQAs. These CQAs
on the drug products’ resistance to changes in their properties also include the impact of raw material variability on the
over the time. Changes in drug product properties are associ- quality of the finished product. This warrants an increased
ated with various input parameters. These include variability excipient-supplier involvement in the formulation development
in the API, the excipients, and the process parameters.19–21 The process. Traditionally, such involvement was either nonexistent

Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24299


REVIEW 3

or limited. Excipients have the potential to demonstrate a vari- coefficient values, ribbon tensile strength, etc.) and final prod-
ety of functionalities depending on the involved application, for uct (e.g., tablet breaking force) properties. However, the excip-
example, food versus pharma, solid versus liquid dosage forms, ient particle size range examined was not sufficiently large to
and so on. They are generally treated by excipient suppliers as adversely influence the immediate-release dosage form CQAs
mere commodities “use as you choose” rather than specialized studied.
product components. Therefore, preformulation scientists are Rudnic et al.29 evaluated the effect of variation in the degree
often unaware of the excipient-supplier’s capabilities to manu- of cross-linkage and extent of carboxymethylation on the disin-
facture and supply excipients meeting their requirements. tegration and dissolution properties of sodium starch glycolate.
Excipients typically constitute a significantly higher fraction The study results showed a substantial modification of disin-
of a formulation than the API. Consequently, the heterogeneous tegrant properties as a function of relatively small changes in
nature of excipient primarily influences the final product at- molecular structure of sodium starch glycolate. The study also
tributes. Regardless of this fact, excipients are relatively less highlighted the importance of specifications for commercially
well characterized. This limitation is now being recognized. available disintegrants.
QbD emphasized the role of excipients as that of enabling the
API to be converted into a pharmaceutical product rather than
mere inert ingredients. Several agencies, including the Inter-
LOT-TO-LOT/BATCH-TO-BATCH VARIABILITY
national Pharmaceutical Excipients Council (IPEC), the Inter-
national Society of Pharmaceutical Engineering (ISPE), the All excipients incorporated in pharmaceutical drug products
American Society for Testing and Materials (ASTM), and the are subjected to a fairly detailed chemical analysis in order to
National Institute for Pharmaceutical Technology and Educa- establish their purity as a part of the quality control protocol.
tion (NIPTE) have focused on excipient characterization.24–26 However, recently, it has been recognized that source-to-source
variability and batch-to-batch variability in the solid state and
the material properties of both the drugs and the excipients
can have profound effects on the final product performance.30–32
EXCIPIENT VARIABILITY AND PRODUCT/PROCESS
Batch-to-batch variation in the excipient properties and varia-
ROBUSTNESS
tion between lots obtained from different suppliers can lead to
Product Quality Research Institute (PQRI) defines process ro- processing challenges or changes in the CQAs of the finished
bustness as the ability of a manufacturing process to tol- products. Such variability can be attributed to the modifica-
erate the expected variability of raw materials, operating tion of the excipient manufacturing process, or adaptation of
conditions, process equipment, environmental conditions, and a more economic source of the raw material to manufacture
human factors.27 Moreton22 similarly described a robust formu- an excipient. However, more subtle variability in the excipi-
lation as the one being able to accommodate the typical vari- ents can occur without the indicated process changes. This can
abilities observed in the “API, excipients, and process” without still present problems for the formulators and the production
compromising the manufacture, stability, or performance of the units.33 Chemically and physically equivalent excipient batches
product. The risk of nonrobust product or process may become may exhibit variable processing performance. Studies carried
significantly higher, if the inherent excipient variability is over- out by Ticehurst et al.34 on "-lactose showed that minor vari-
looked, missed, or even underestimated. A detailed knowledge ations in the degree of surface crystallinity may result in lot-
of the excipient manufacturing history or the supplier process to-lot variability. The authors measured the surface energies of
capability may reduce such a risk. Pharmaceutical excipients four physically and chemically equivalent batches of "-lactose
are typically manufactured at a large-scale using some form of obtained from the same source. They used the absorption be-
a continuous or a batch process. Manufacturing processes are havior of a range of polar and nonpolar probes in inverse gas
designed to produce material within a relatively broad range chromatography. The results showed minor differences in the
of specifications. The manufactured excipient will inevitably surface energies of lactose from different lots. These differences
show some variation within this range. The robustness of the were attributed to minor variations in the crystallinity and
final drug product is dependent on the ability of the formula- purity of the batches. Martino et al.35 evaluated the lot-to-lot
tion and the process to accommodate this variability without variability in the compression behavior of seven lots of lactose
compromising the final product performance.21 This is the core monohydrate with an aim of identifying the cause of variability.
of the “Design Space” concept utilized in QbD. Incorporating They had hypothesized four potential causes to explain lot-to-
excipient variability into the design of experiments, used to es- lot variability in the selected batches of lactose monohydrate.
tablish the “design space” and possibly the “edge of failure” can These causes were the differences in particle size distribution,
produce a robust formulation.24 presence of anhydrous lactose, presence of $-lactose, and varia-
Independent studies have been carried out over the years tion in the crystalline behavior. The study results showed that
to identify and quantify the influence of excipient variabil- the particle size distribution was not significantly different be-
ity on their functionality and eventually on the final product tween lots. The presence of anhydrous lactose and $-lactose,
CQAs. Kushner et al.28 studied the impact of the excipient although observed, was not found in quantities significant to
material property variations for three common excipients, mi- make an impact. The degree of the order of crystalline struc-
crocrystalline cellulose (MCC), spray-dried lactose, and mag- ture was found to be the main reason for the observed lot-to-lot
nesium stearate on the performance and manufacturability of variation. Landin et al.36 studied the influence of particle size
an immediate-release, dry-granulated, solid oral dosage forms. on the dehydration behavior of dicalcium phosphate dihydrate.
These studies showed that the variations in the MCC and the Differences in the particle size were found to be responsible for
lactose particle size led to statistically significant changes in the interlot and intermanufacturer variability observed in di-
the intermediate (e.g., blend particle size, blend flow function calcium phosphate dihydrate. Batch-to-batch differences in the

DOI 10.1002/jps.24299 Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES


4 REVIEW

excipient properties has been shown to influence a broad range The increasing sophistication of the processing equipment
of excipient functionalities used in the solid, semisolid, and liq- poses higher demands on the excipient quality than ever be-
uid dosage formulations.37–40 Alvarez-Lorenzo et al.37 compared fore. Uncontrolled variability in the excipient quality may lead
the physicochemical properties of three lots of high-viscosity to inefficiencies during the development and production pro-
hydroxypropylcellulose (HPC). The results showed clear differ- cess. This can also produce unacceptable variability in the end-
ences in molecular weight, molecular structure, particle size product quality. Therefore, adequate consistency of an excip-
distribution, particle shape, and water affinity for HPC lots ient property is essential to obtain a constant quality of the
from the same supplier, as well as lots from different suppli- final drug product and to facilitate a smooth production. The
ers. The study also demonstrated differences in the drug re- batch-to-batch variations of excipients within specification can
lease behavior of the dosage forms prepared using these lots of be accommodated by good formulation practice. This includes
HPC. The authors hypothesized that lot-to-lot variation in the the use of different batches of inactive constituents during the
physicochemical properties of HPC influences the compression development and scale up. It might be a good idea to use the
behavior and the rate and extent of water uptake by the dosage batches of critical excipients at the outer limits of their specifi-
forms. This was attributed to the resulting observed variability cations during the development stage.
in the drug release. O’Laughlin et al.39 studied the influence
of lot-to-lot variation in the physicochemical properties of glyc-
eryl monostearate on the stability of an oil-in-water cream. The
SOURCE-TO-SOURCE VARIATION
difference found in the “acid value” of glyceryl monostearate
lots was a major determinant of the stability of the prepared Different suppliers are most likely the cause of variability in
cream. Lots with higher “acid value” demonstrated better sta- excipient batches. Excipients may vary in quality because of dif-
bility. Perez Marcos et al.40 evaluated seven lots of Carbopol R
ferences at the molecular and macroscopic level such as impu-
934, which is a high and a heterogeneous molecular weight rity profile, molecular weight distribution (in case of polymers),
acrylic acid-based cross-linked polymer, commonly used as an polymorphic form, particle morphology, particle size distribu-
extended-release matrix former in tablets. No significant differ- tion, surface properties, hydration state, moisture content, and
ences in the infrared profiles, density, or carboxylic acid content so on.43
between the lots were reported. However, the rheological prop- A large group of pharmaceutical excipients is derived from
erties of the aqueous dispersions were significantly different natural products. This increases the excipient’s susceptibility
between the lots. This difference can be because of the differ- to seasonal as well as natural and artificial changes affecting
ences in the mean molecular weight between the polymer lots. the end characteristics of the excipient.44,45 A given excipient
Maincent41 summarized that although excipients are used to may be procured from a number of different manufacturers. It
prepare dosage forms to meet official pharmacopoeial mono- is desirable for a pharmaceutical product to have more than
graph specifications, their properties may vary from batch to one source for each raw material because of the constant sup-
batch. These variations can alter the physicochemical proper- ply requirements. Excipients obtained from different suppli-
ties, as well as functionalities of the final dosage formulations. ers are often interchanged in the formulation. However, ex-
Their study showed that the tablets prepared with the same cipients of similar grade from different manufacturers often
excipient (e.g., MCC) originating from several suppliers might show variability in their properties. Haware et al.46 carried
display dramatic differences in their functionalities. The main out a qualitative and quantitative analysis of the compression
difference was associated with the in vitro dissolution rate, behavior of similar grades of MCC using multivariate meth-
which may consequently affect the bioavailability. In a recently ods. They used Avicel PH 101 (FMC Biopolymer, Brussels,
R

published article, Kushner20 proposed a simple method of using Belgium) and Vivapur 101 (JRS Pharma, Rosenberg, Deutsch-
R

the quantitative certificate of analysis (CoA) data to assess the land, Germany) as representative grades obtained from differ-
extent of lot-to-lot variability in excipients during drug product ent vendors.46 The data obtained from the principle component
development process. This study retrospectively carried out a analysis (PCA) and the partial least squares (PLS) analysis was
principal component analysis (PCA)-based multivariate anal- able to demonstrate the variability in the physical and compres-
ysis of quantitative physical-chemical properties found in the sion properties of MCC obtained from different sources (Fig. 2).
CoAs of several commonly used tableting excipients such as Parker and coworkers47–50 demonstrated the effect of the
MCC, spray-dried lactose, and magnesium stearate. A large source variation on the rheological behavior during the wet
number of CoAs for each material, that is, 968 for MCC, 439 granulation of two MCCs using rheological profiles in a series of
for spray-dried lactose, and 168 for magnesium stearate, were comprehensive studies. They also showed that dissimilar poly-
obtained from individual vendors and analyzed. The analysis mers interact differently with cellulose substrates during the
of the CoAs showed that although the lots of excipients were wet massing process. In addition, for a second cellulose mate-
manufactured within the vendor’s specifications, the PCA was rial, variations were also observed in the pattern of interaction
able to detect minor, site-to-site, and year-to-year variability in between the binders and the excipient.
these excipients. These examples demonstrate a strong need Landin et al.51 demonstrated the differences in the prop-
for an adequate control of variability, even for the excipients erties of MCC obtained from different source materials (wood
procured from the same manufacturer. The IPEC has drafted pulp) and different processing methods. These differences did
the significant change guide for bulk pharmaceutical excipi- not significantly influence tableting parameters but were sug-
ents. This guide directs the excipient manufacturer to evaluate gested to be the potential causes of variability in the final phar-
the significance of changes involving the manufacture of bulk maceutical products’ functionalities.51 Landin et al.45 also stud-
pharmaceutical excipients. This guide also steers to determine ied the characteristics of prednisone tablets prepared by direct
the need for informing excipient customers and regulatory au- compression with six previously characterized MCCs from vari-
thorities about the nature of the change.42 ous sources. MCCs of similar particle size produced tablets with

Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24299


REVIEW 5

intramanufacturer variability among the grades and lots of


xanthan gum was reported in this study. However, this vari-
ability was not reflected in the NF viscosity test specifications.
Whiteman and Yarwood evaluated six lactose-based mate-
rials as direct compression tablet excipients. The studied lac-
toses showed changes in the compaction properties to varying
degrees.58 These studies pointed out that although exchang-
ing one excipient for another in an existing formulation may
be economical, compendial specifications cannot adequately
differentiate between products with differing compaction
properties.

INFLUENCE ON FUNCTIONALITY
The excipients are included in the formulation to improve the
drug products’ processability, and to meet the required end-
Figure 2. The PCA score plots of the physical and compression prop- product specifications. The desirable excipient properties relate
erties of AvicelR PH 101 and VivapurR 101. The variables explain the to the functional performance or the functionality. Excipient
two principle components (PCs). The two displayed PCs explained 49% functionalities are qualitative classifications describing the ra-
and 21% of the variance, respectively. YPpl, yield pressure of plastic tionale or purposes or roles of an excipient inclusion in a drug
deformation; YPelI , yield pressure of immediate elastic recovery; WoC,
product formulation. Excipient functionality has been defined
work of compression; WoEI , work of immediate elastic recovery; d50,
mean particle size; HF, Hausner’s flowability ratio; and TS, tablet ten-
in the IPEC Excipient Qualification Guide as: “A desirable prop-
sile strength. Reproduced from Haware et al.46 with permission from erty of an excipient that aids manufacturing and improves the
Pharmaceutical Development and Technology. manufacture, quality or performance of the drug product.”59,60
On the contrary, excipient performance is a broader term en-
compassing the excipient properties, including functionalities,
similar mechanical and microstructural properties. In contrast, in a specific formulation.59,61
the dissolution rate of prednisone varied significantly with the The main questions regarding the excipient functionality
particle size and the chemical composition of MCC.45 are: how to assess and control it. Realistically, functionality can
The current national formulary (NF) monographs do not pro- only be assessed properly in the context of the finished phar-
vide the tests reflecting the disintegrant functionality of sodium maceutical product. Moreover, each application (formulation)
starch glycolates. Thus, one cannot assume reliable perfor- has unique requirements for functionality. The role of pharma-
mance of the disintegrants from different sources meeting NF copoeias in the assessment of excipient functionality is a topic
standards.52 Bolhuis et al.53 demonstrated the differences in of current debate. There appears to be a general agreement that
the disintegration time of tablets containing sodium starch gly- excipient monographs have not addressed excipient functional-
colate as a disintegrant. The sodium starch glycolate included ity or performance per se. Clearly, it was not the original intent
in the formulations were obtained from starch of different ori- of the excipient monographs. The US and Europe are adopting
gins. The differences in the rate of penetration of water into two very different approaches. The European Pharmacopoeia
the particles of sodium starch glycolate were attributed to the has introduced a nonmandatory functionality related charac-
differences in the chemical composition, crystallinity, and par- teristics (FRCs) sections in certain excipient monographs. This
ticle size of the starches used in the manufacture of sodium is not an option in the US. Legally, all sections of a monograph
starch glycolates. Shah and Augsburger54 carried out similar in both the USP, and the NF are mandatory.16,24,60 The current
studies to characterize the physical properties of sodium starch regulatory environment including the QbD paradigm goes be-
glycolate obtained from various sources. Differences in particle yond simply identifying an excipient function. It emphasizes
sizes, surface area, porosity, surface morphology, and viscos- excipient performance through the identification, evaluation,
ity between sources of sodium starch glycolate resulted in the and control of critical material attributes (CMAs) that assure
differences in the extent of liquid uptake by the disintegrant. consistent performance throughout a product’s life cycle.59 The
Landin et al.55 evaluated the characteristics of four brands development of appropriate functionality tests has been ham-
of MCC manufactured in Finland, India, Ireland, and Japan. pered by numerous difficulties and complexities, including a
They found that the brands differed significantly in the chemi- lack of a precise understanding of the mechanisms of excipient
cal composition, crystal structure, and particle size of MCC. All functionality.
these differences were potentially relevant to the differences in Zhao and Augsburger62 carried out a comprehensive study
the powder flow properties and rheological behavior. Gamble on the function-related properties such as particle size, intrin-
et al.56 quantified batch-to-batch and vendor-to-vendor varia- sic and bulk swelling, water uptake ability, and the influence of
tions in the solid-state characteristics of multiple batches of medium pH on these properties on five brands of croscarmellose
anhydrous lactose obtained from three vendors. The study re- sodium. Studies specially emphasized on developing a discrim-
ported the subsequent impact of these differences on process inating model formulation for the disintegrants performance
ability and/or functionality.56 Thacker et al.57 investigated the comparison and the quality control purposes.62 These studies
intermanufacturer and intramanufacturer variability among indicated that particle size, total degree of substitution, and the
the different grades and lots of xanthan gum produced ratio of basic to acidic substituents were among the most im-
by two manufacturers. A significant intermanufacturer and portant factors affecting the disintegrants swelling efficiency.62

DOI 10.1002/jps.24299 Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES


6 REVIEW

Figure 3. Analysis of excipient variability.

Rojas et al.63 studied the influence of the nature of MCC, diameter and the use of talc in association with magnesium
MCC I (prepared by acid hydrolysis of wood pulp) and MCC II stearate as two potential solutions to reduce the impact of
(prepared by mercerization of MCC I) on the physical properties the variability of magnesium stearate. Recently, our labora-
of tablets. Differences in the physical properties of MCC I and tory studied the lubrication functionality of different lots of
MCC II dictated the powder and the tableting properties. These bovine- and vegetable-derived magnesium stearates as a func-
findings emphasized the importance of selecting the right MCC tion of molecular and macroscopic properties using multivari-
grade before formulating a drug in a solid dosage form. Shah ate analysis methods.68 These studies showed that molecular
et al. studied relationships of physical properties differences properties such as crystallinity, moisture content, and macro-
with the crospovidones functionality obtained from multiple scopic properties such as particle size and specific surface area
sources.64 The study observed major differences in the physi- are mainly responsible for magnesium stearate lubrication
cal characteristics of crospovidone NF obtained from various efficiency.68
sources. These variations resulted in differences in the tablet The USP distinguishes four different HPMC grades by the
disintegration and dissolution containing insoluble crospovi- proportions of methoxy and hydroxypropoxy groups.69 The
done. Chamarthy et al.65 illustrated the influence of raw mate- properties of individual grades will furthermore depend on
rial variability on the excipient’s functionality during process- their molecular weight. However, it has been demonstrated
ing. Studies observed that differently processed starch samples that batches obtained from different suppliers of a similar
had essentially similar physical characteristics such as particle pharmacopoeial HPMC grade can lead to significantly differ-
size, specific surface area, and apparent crystallinity, and so on. ent performance.70
On the contrary, the compaction properties of the two lots were
distinctly different tested under all environmental and process-
ing conditions. Clearly, the two lots are diverse materials as per
EXCIPIENT VARIABILITY ANALYSIS
the functionality point of view. The differences were attributed
to the altered surface energetic properties of the excipient. Excipient variability may significantly affect the physicochem-
Zhao and Augsburger66 observed differences in the disin- ical properties and the excipients functionality in final dosage
tegration efficiency of the three classes of superdisintegrants. forms. Therefore, it is important to have analytical methods
The study concluded that the three disintegrants represent- in place for accurately assessing and identifying differences in
ing the three main classes of superdisintegrants (Ac-Di-Sol , R
these materials.23
Primojel , and Polyplasdone XL10) differed in their ability to
R R
Figure 3 represents the different approaches used to assess
disintegrate model tablets into their primary particles when excipient variability at molecular and macroscopic level. Sev-
used at the same percentage by weight fractions. The study eral analytical techniques have been developed over the years
further concluded that such differences could potentially affect to accurately analyze and identify the excipient variability at
drug dissolution. Barra and Somma67 estimated the physico- a molecular level. Some of the commonly utilized sophisti-
chemical property variation in thirteen commercial batches of cated analytical techniques include moisture sorption analyzer,
magnesium stearate from three vendors using various physico- differential scanning calorimetry/thermogravimetric analysis,
chemical tests. They found that the physical properties of mag- powder X-ray diffractometry, near-infrared spectroscopy, and
nesium stearate varied between vendors as well as between solid-state nuclear magnetic resonance (SSNMR). Vogt and
lots obtained from the same vendor. The study proposed uti- Williams in a recent paper-reviewed several advanced ap-
lization of a standardized magnesium stearate mean particle proaches for effective solid-state analysis of pharmaceuticals.71

Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24299


REVIEW 7

Excipient Variability Analysis at a Molecular Level regressions, principle component regressions, and PLS
regressions).91 Haware et al.89,92,93 utilized multivariate meth-
A molecular level excipient characterization such as poly-
ods to differentiate the variability in the functionalities of var-
morphic form identification, structural differences, crys-
ious grades of directly compressible celluloses and lactoses, as
talline/amorphous content, and water content variations can
well as lubrication efficiencies of varying sources of magnesium
be detected using SSNMR spectroscopy experiments. SSNMR
stearate.
spectroscopy can be very useful during the preformulation
and the formulation stages. This is particularly important
when the source of an excipient changes, or the manufactur-
ing and/or storage conditions could potentially alter the excip- QbD AND EXCIPIENT VARIABILITY
ient functionalities.72,73 Sperger and Munson74 demonstrated
Managing excipient variability is an essential element in the
the applicability of SSNMR in detecting the differences in the
designing and the manufacturing of a robust drug product. It
structural properties among common polysaccharide-based ex-
is an integral task in applying QbD principles.26 The US FDA’s
cipients in a recent study.
21st century cGMP initiative and ICH Q8 guidelines encour-
Near-infrared spectroscopy is another analytical technique
age the pharmaceutical excipient users to apply QbD princi-
gaining popularity for analyzing excipient variability and
ples in developing “flexible” formulations and processes. This
functionality.10,75–81 Gabrielsson et al.82 used NIR to quantify
is necessary to cope with anticipated raw material variability
the amount of lot-to-lot variability and its impact on the per-
in producing robust and consistently performing products.24,94
formance of a new drug product. NIR was found to be useful in
One approach to achieve this could be to optimize and control
confirming the robustness of their product in the face of exist-
the process based on the raw material properties. Such process
ing excipient lot-to-lot variability. It is important to understand
control can compensate the incoming variability to yield the fin-
the capabilities and limitations of the utilized technique while
ished product, consistently meeting the quality target product
choosing a technique for the excipient variability analysis. In-
profile (QTPP). Another approach is to specify the raw materi-
strumental effects (instrument drift and aging, lamp changes,
als’ CQAs tightly enough to ensure a consistent performance to
etc.), environmental effects (changes in relative humidity in
the QTPP. Of these approaches, the first approach dealing with
storage facilities), and active ingredient(s) and excipients vari-
adjustments during the manufacturing process to compensate
ability are likely to affect the analytical abilities of a technique.
for inherent raw material variability appears to be more viable.
Igne et al.83 studied the effects of instrumental aging, excipient
A fundamental concept within QbD is the design space. ICH
particle size distribution, excipient manufacturer variability,
Q8 defines design space as follows: the multidimensional com-
and active ingredient physical transformation. The study con-
bination and interaction of input variables (e.g., material at-
cluded that variation in the raw material properties had a sig-
tributes) and process parameters that have been demonstrated
nificant impact on the precision and the accuracy of predictions
to provide assurance of quality. Working within the design
from the model.
space is not considered as a change. Movement out of the design
Excipient Variability Analysis at Macroscopic Level space is considered to be a change and would normally initiate
a regulatory postapproval change process. Design space is pro-
The macroscopic excipient properties such as particle morphol- posed by the applicant and is subject to regulatory assessment
ogy, and density (bulk, tapped, and particle) can be measured and approval.24
by scanning electron microscopy and other official methods Quality-by-Design is a relatively new approach in the US.
mentioned in the USP.5,6 These measurements are used for an Correct applications of QbD allow industry to adopt better,
indirect assessment of the variations in the excipient charac- quicker, and more efficient working practices. This encourages
teristics. In addition to these compendial methods, the noncom- development of the robust formulations and processes.16 IPEC
pendial methods such as powder rheometry has been used to Americas has formed a QbD product development committee to
evaluate the powder flowability of excipients.84 The variabil- address the following areas: proper selection and use of excipi-
ity in excipient compressibility can be assessed with sophis- ent performance tests (addressing functionality)—decision tree
ticated instruments such as the compaction simulator.4,9,85,86 approach; robust formulations development (including QbD,
Compaction simulators allow simulation of the controlled com- PAT, etc.); and the introduction of coprocessed excipients with
pression parameters similar to industrial manufacturing con- customized functionalities (removal of regulatory barriers and
ditions. This technique produces a highly accurate, real-time customer acceptance).16,60 QbD concepts emphasize the need
force-displacement data with a small sample-size requirement for characterizing CMAs (e.g., micromeritic, chemical, thermal,
of the materials. The parameterization of the obtained force- rheological, and mechanical properties) and define their role
displacement data with well-known global mathematical equa- in the formulation and manufacturing processes. QbD concepts
tions such as Heckel, Kawakita, or other energy descriptors also help in understanding the role of excipients in the for-
allow the evaluation of material’s deformation behavior.14,87,88 mulation and the process using the science and risk-based ap-
This approach has been commonly utilized to identify the vari- proach for excipient selection (compatibility and functionality)
ability in the deformation tendency of the excipients.89,90 and improved excipient characterization. QbD provides a clear
understanding of the tolerance limit of the excipient variability
Excipient Variability Analysis using Multivariate Techniques
by a process, to ensure a predictable, high-quality, reproducible,
Characterization of excipient variability at a molecular and and stable product. In addition, QbD defines relevant analytical
macroscopic level, using above-mentioned sophisticated instru- tools to assess the excipient variability impact on the formula-
mentation, generates a huge amount of data. Multivariate tion, process, and product performance.24,26
analysis methods can be used to quantify excipient variability Several researchers have now begun to incorporate
both qualitatively (PCA) and quantitatively (multiple linear QbD principles for the characterization of excipient

DOI 10.1002/jps.24299 Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES


8 REVIEW

variability. Fu et al.95 determined the intergrade and interbatch 12. Cantor SL, Khan MA, Gupta A. 2014. Development and opti-
variability of sodium alginate using appropriate rheological mization of taste-masked orally disintegrating tablets (ODTs) of clin-
methods and conditions. This provided insight into the delin- damycin hydrochloride. Drug Dev Ind Pharm 7:1–9.
eation of the design space as a part of QbD for sodium alginate- 13. Kharb V, Saharan VA, Jadhav H, Purohit S. 2014. Formulation and
evaluation of lipid based taste masked granules of ondansetron HCl.
based formulations.95
Eur J Pharm Sci 62:180–188.
14. Li X, Guo Z, Hao JB, Li B, Liu CB, Guo T, Li HY, Shi SL,
Wang LY, Zhang JW. 2014. Synergetic taste masking of lipid coat-
CONCLUSIONS ing and beta-cyclodextrin inclusion. Yao Xue Xue Bao 49(3):392–
398.
The use of excipients in drug products is ubiquitous and in- 15. Yuan J, Liu T, Li H, Shi T, Xu J, Liu H, Wang Z, Wang Q, Xu
dispensable. An unintentional variability in the excipients is L, Wang Y, Li S. 2014. Oral sustained-release suspension based on a
unavoidable. Excipients batch-to-batch, source-to-source, and novel taste-masked and mucoadhesive carrier-ion-exchange fiber. Int
supplier-to-supplier variability can be present at both molecu- J Pharm 472(1–2):74–81.
lar as well as macroscopic levels. Thus, clear understanding and 16. Moreton RC. 2010. Functionality and performance of excipients in
quantification of the nature, source, and the extent of excipient a Quality-by-Design world: Part I. In Functionality and performance
variability is critical for robust formulation development. It is of excipients in a Quality-by-Design world; Moreton C, Ed. Russell
very important to understand the impact of these variabilities Publishing (American Pharmaceutical Review), Brasted Kent: UK.
17. Nachaegari SK, Bansal AK. 2004. Coprocessed excipients for solid
on the excipient functionality and eventually on the CQAs of
dosage forms. Pharm Tech 52–64.
the final product. An intelligent use of QbD and design space
18. Hebestreit P. 2009. Addressing specific regulatory excipient re-
principles helps understand this impact of excipient variability quirements in the marketing authorization. Nice: The International
on its functionality and performance. The application of multi- Pharmaceutical Excipients Council Europe.
variate analysis with sophisticated instrumentation serves as 19. Belic A, Skrjanc I, Bozic DZ, Vrecer F. 2010. Tableting process
important tools for the characterization and quantification of optimisation with the application of fuzzy models. Int J Pharm 389(1–
excipient variabilities. 2):86–93.
20. Kushner JT. 2013. Utilizing quantitative certificate of analysis data
to assess the amount of excipient lot-to-lot variability sampled during
drug product development. Pharm Dev Technol 18(2):333–342.
REFERENCES
21. Moreton RC. 2010. Functionality and performance of excipients
1. Haywood A, Glass BD. 2011. Pharmaceutical excipients—Where do in a Quality-by-Design world: Excipient variability, QbD, and robust
we begin? Aust Prescr 34:112–114. formulations. Am Pharm Rev 2009(12):24–27.
2. Gstirner F, Pick C. 1967. Effect of Aerosil and “filling material R 22. Moreton RC. 2006. Functionality and performance of ex-
972” on the flow properties of zinc oxide. Arch Pharm Ber Dtsch Pharm cipients. Pharm Technol. Accessed September 11, 2014, at:
Ges 300(6):504–515. http://www.pharmtech.com/functionality-and-performance-excipients.
3. Abe H, Yasui S, Kuwata A, Takeuchi H. 2009. Improving powder flow 23. Moreton RC. 2009. Functionality and performance of excipients in
properties of a direct compression formulation using a two-step glidant a Quality-by-Design world Part 4: Obtaining information on excipient
mixing process. Chem Pharm Bull 57(7):647–652. variability for formulation design space. Am Pharm Rev 2009(12):28–
4. Garzon Mde L, Villafuerte L. 2002. Compactibility of mixtures of cal- 32.
cium carbonate and microcrystalline cellulose. Int J Pharm 231(1):33– 24. USFDA. 2009. Guidance for industry—ICH Q8 (R2) pharmaceutical
41. development. In CDER C, Ed. Silver Spring, Maryland: USFDA, pp 1–
5. Nokhodchi A, Maghsoodi M, Hassan-Zadeh D, Barzegar-Jalali M. 20.
2007. Preparation of agglomerated crystals for improving flowability 25. Winkle HN. 2007. Implementing Quality by Design. In CDER, Ed.
and compactibility of poorly flowable and compactible drugs and excip- Silver Spring, Maryland: USFDA.
ients. Powder Technol 175(2):73–81. 26. Carlin B, Moreton C. 2010. Flexible approaches to accommodate ex-
6. Vromans H, Lerk CF. 1988. Densification properties and compactibil- cipient variability using the principles of QbD. Pharm Technol (34):s36–
ity of mixtures of pharmaceutical excipients with and without magne- s39.
sium stearate. Int J Pharm 46(3):183–192. 27. Glodek M, Liebowitz S, McCarthy R, McNally G, Oksanen C,
7. Derganc J, Likar B, Bernard R, Tomaževič D, Pernus F. 2003. Real- Schultz T, Sundararajan M, Vorkapich R, Vukovinsky K, Watts C,
time automated visual inspection of color tablets in pharmaceutical Millili G. 2006. Process robustness—A PQRI white paper. Pharm Eng
blisters. Real-Time Imaging 9(2):113–124. 26(6):1–11.
8. Dave VS, Fahmy RM, Bensley D, Hoag SW. 2012. EudragitR 28. Kushner JT, Langdon BA, Hiller JI, Carlson GT. 2011. Exam-
RS PO/RL PO as rate-controlling matrix-formers via roller com- ining the impact of excipient material property variation on drug
paction: Influence of formulation and process variables on functional product quality attributes: A quality-by-design study for a roller
attributes of granules and tablets. Drug Dev Ind Pharm 38(10):1240– compacted, immediate release tablet. J Pharm Sci 100(6):2222–
1253. 2239.
9. Dave VS, Fahmy RM, Hoag SW. 2013. Investigation of the physical- 29. Rudnic EM, Kanig JL, Rhodes CT. 1985. Effect of molecular struc-
mechanical properties of EudragitR RS PO/RL PO and their mix- ture variation on the disintegrant action of sodium starch glycolate. J
tures with common pharmaceutical excipients. Drug Dev Ind Pharm Pharm Sci 74(6):647–650.
39(7):1113–1125. 30. Butcher AE, Jones TM. 1972. Some physical characteristics of mag-
10. Dave VS, Fahmy RM, Hoag SW. 2014. Near-infrared spectroscopic nesium stearate. J Pharm Pharmacol 24:1P–9P.
analysis of the breaking force of extended-release matrix tablets pre- 31. Holzer AW, Sjogren J. 1981. Evaluation of some lubricants by the
pared by roller-compaction: Influence of plasticizer levels and sintering comparison of friction coefficients and tablet properties. Acta Pharm
temperature. Drug Dev Ind Pharm.[Epub ahead of print] Suec 18(3):139–148.
11. Williams RO, 3rd, Reynolds TD, Cabelka TD, Sykora MA, 32. Rees JE, Rue PJ. 1978. Interbatch variation in compression prop-
Mahaguna V. 2002. Investigation of excipient type and level on drug erties of Elcema G250 [proceedings]. J Pharm Pharmacol 30:25P.
release from controlled release tablets containing HPMC. Pharm Dev 33. York P. 1983. Solid-state properties of powders in the formulation
Technol 7(2):181–193. and processing of solid dosage forms. Int J Pharm 14(1):1–28.

Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24299


REVIEW 9

34. Ticehurst MD, York P, Rowe RC, Dwivedi SK. 1996. Characteri- 53. Bolhuis GK, Van Kamp HV, Lerk CF. 1986. On the similarity of
sation of the surface properties of A-lactose monohydrate with inverse sodium starch glycolate from different sources. Drug Dev Ind Pharm
gas chromatography, used to detect batch variation. Int J Pharm 141(1– 12(4):621–630.
2):93–99. 54. Shah U, Augsburger L. 2002. Multiple sources of sodium starch
35. Martino PD, Martelli S, Guyot-Hermann AM, Guyot JC, Drache glycolate, NF: Evaluation of functional equivalence and development
M, Conflant P. 1993. The batch-to-batch non-reproducibility of the of standard performance tests. Pharm Dev Technol 7(3):345–359.
compression ability of lactose reasons and detection. STP Pharma Sci 55. Landin M, Martinez-Pacheco R, Gomez-Amoza JL, Souto C,
3(6):436–441. Concheiro A, Rowe RC. 1993. Effect of country of origin on the
36. Landin M, Rowe RC, York P. 1994. Particle size effects on the properties of microcrystalline cellulose. Int J Pharm 91(2–3):123–
dehydration of dicalcium phosphate dihydrate powders. Int J Pharm 131.
104(3):271–275. 56. Gamble J, Chiu W-S, Gray V, Toale H, Tobyn M, Wu Y. 2010. In-
37. Alvarez-Lorenzo C, Castro E, Amoza G, Luis J, Martinez-Pacheco vestigation into the degree of variability in the solid-state properties of
R, Souto C, Concheiro A. 1998. Intersupplier and interlot variability common pharmaceutical excipients—Anhydrous lactose. AAPS Pharm
in hydroxypropyl celluloses: Implications for theophylline release from Sci Tech 11(4):1552–1557.
matrix tablets. Pharm Acta Helv 73(2):113–120. 57. Thacker A, Fu S, Boni R, Block L. 2010. Inter- and intra-
38. Dunn RL, English JP, Strobel JB, Cowsar DR, Tice TR. 1988. manufacturer variability in pharmaceutical grades and lots of xanthan
Preparation and evaluation of lactide/glycolide copolymers for drug gum. AAPS Pharm Sci Tech 11(4):1619–1626.
delivery. In Polymers in medicine III; Migliaresi C, Ed. Ams- 58. Whiteman M, Yarwood RJ. 1988. The evaluation of six lactose-based
terdam, The Netherlands: Elsevier Science Publishers, pp 149– materials as direct compression tablet excipients. Drug Dev Ind Pharm
160. 14(8):1023–1040.
39. O’Laughlin R, Sachs C, Brittain H, Cohen E, Timmins P, Varia 59. Carlin B. 2012. Quality risk management of compliant excipients.
S. 1989. Effects of variations in physicochemical properties of glyceryl J Excip Food Chem 3(4):143–153.
monostearate on the stability of an oil-in-water cream. J Soc Cosmet 60. IPEC. 2008. Qualification of excipients for use in pharmaceuti-
Chem 40(4):215–229. cals. Accessed September 12, 2014, at: http://ipecamericas.org/sites/
40. Perez-Marcos B, Martinez-Pacheco R, Gomez-Amoza JL, Souto C, default/files/ExcipientQualificationGuide.pdf. Arlington, Virginia:
Concheiro A, Rowe RC. 1993. Interlot variability of carbomer 934. Int IPEC-Americas.
J Pharm 100(1–3):207–212. 61. Sheehan C, Amidon GE. 2011. Compendial standards and excipi-
41. Maincent P. 1999. The interchangeability of excipient formulations ent performance in the QbD era: USP excipient performance chapter
and the eventual consequences. Therapie 54(1):5–10. <1059>. Am Pharma Rev 14(6):10–18.
42. Goode SA, Carter D, Jacobs MG, Klug DB, Merrell PH, Moreton RC, 62. Zhao N, Augsburger LL. 2006. The influence of product brand-
Schoneker DR, Silverstein IB, Ulman KL, Meter AV, Zawislak P. 2009. to-brand variability on superdisintegrant performance. A case
Significant change guide for bulk pharmaceutical excipients IPEC- study with croscarmellose sodium. Pharm Dev Technol 11(2):179–
Americas. Accessed September 12, 2014, at: http://ipecamericas.org/ 185.
sites/default/files/IPECSignificantChangeGuide2009.pdf. 63. Rojas J, Lopez A, Gamboa Y, Gonzalez C, Montoya F. 2011. As-
43. Matthews BR. 2009. Drug product—Excipients. In International sessment of processing and polymorphic form effect on the powder and
pharmaceutical product registration; Cartwright AC, Matthews BR, tableting properties of microcrystalline celluloses I and II. Chem Pharm
Eds. 2nd ed. Informa Healthcare, London: UK, pp 234–247. Bull 59(5):603–607.
44. Landin M, Gonzalez MP, Souto C, Concheiro A, Gomez-Amoza JL, 64. Shah U, Augsburger L. 2001. Evaluation of the functional equiva-
Martinez-Pacheco R. 1993. Comparison of two varieties of microcrys- lence of crospovidone NF from different sources. I. Physical character-
talline cellulose as filler-binders II. Hydrochlorothiazide tablets. Drug ization. Pharm Dev Technol 6(1):39–51.
Dev Ind Pharm 19(10):1211–1220. 65. Chamarthy S, Pinal R, Carvajal MT. 2009. Elucidating raw ma-
45. Landin M, Vazquez MJ, Souto C, Concheiro A, Gomez-amoza JL, terial variability: Importance of surface properties and functional-
Martinez-pacheco R. 1992. Comparison of two varieties of microcrys- ity in pharmaceutical powders. AAPS Pharm Sci Tech 10(3):780–
talline cellulose as filler-binders I. Perdnisone tablets. Drug Dev Ind 788.
Pharm 18(3):355–368. 66. Zhao N, Augsburger LL. 2005. Functionality comparison of 3 classes
46. Haware RV, Bauer-Brandl A, Tho I. 2010. Comparative evalua- of superdisintegrants in promoting aspirin tablet disintegration and
tion of the powder and compression properties of various grades and dissolution. AAPS Pharm Sci Tech 6(4):E634–E640.
brands of microcrystalline cellulose by multivariate methods. Pharm 67. Barra J, Somma R. 1996. Influence of the physicochemical variabil-
Dev Technol 15(4):394–404. ity of magnesium stearate on its lubricant properties: Possible solu-
47. Parker MD, Rowe RC. 1991. Source variation in the wet massing tions. Drug Dev Ind Pharm 22(11):1105–1120.
(granulation) of some microcrystalline celluloses. Powder Technol 65(1– 68. Haware RV, Shivagari R, Johnson PR, Staton S, Stagner WC,
3):273–281. Gupta MR. 2014. Application of multivariate methods to evaluate the
48. Parker MD, York P, Rowe RC. 1990. Binder-substrate interactions functionality of bovine- and vegetable-derived magnesium stearate. J
in wet granulation. 1: The effect of binder characteristics. Int J Pharm Pharm Sci 103(5):1466–1477.
64(2–3):207–216. 69. Siepmann J, Peppas NA. 2001. Modeling of drug release from de-
49. Parker MD, York P, Rowe RC. 1991. Binder-substrate interactions livery systems based on hydroxypropyl methylcellulose (HPMC). Adv
in wet granulation. 2: The effect of binder molecular weight. Int J Drug Deliv Rev 48(2--3):139–157.
Pharm 72(3):243–249. 70. Shah AC, Britten NJ, Olanoff LS, Badalamenti JN. 1989. Gel-
50. Parker MD, York P, Rowe RC. 1992. Binder-substrate interactions matrixsystems exhibiting bimodal controlled release for oral drug de-
in wet granulation. 3: The effect of excipient source variation. Int J livery. J Control Release 9(2):169–175.
Pharm 80(1–3):179–190. 71. Vogt FG, Williams GR. 2010. Advanced approaches to effective
51. Landin M, Martinez-Pacheco R, Gomez-Amoza JL, Souto C, solid-state analysis: X-ray diffraction, vibrational spectroscopy and
Concheiro A, Rowe RC. 1993. Effect of batch variation and source of solid-state NMR. Am Pharm Rev 13(7).
pulp on the properties of microcrystalline cellulose. Int J Pharm 91(2– 72. Berendt RT, Sperger DM, Munson EJ, Isbester PK. 2006. Solid-
3):133–141. state NMR spectroscopy in pharmaceutical research and analysis.
52. Shah U, Augsburger L. 2001. Evaluation of the functional equiv- TrAC. Trends Anal Chem 25(10):977–984.
alence of crospovidone NF from different sources. II. Standard perfor- 73. Lubach JW, Xu D, Segmuller BE, Munson EJ. 2007. Investigation
mance test. Pharm Dev Technol 6(3):419–430. of the effects of pharmaceutical processing upon solid-state NMR re-

DOI 10.1002/jps.24299 Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES


10 REVIEW

laxation times and implications to solid-state formulation stability. J 85. Tatavarti AS, Muller FX, Hoag SW. 2008. Evaluation of the de-
Pharm Sci 96(4):777–787. formation behavior of binary systems of methacrylic acid copolymers
74. Sperger DM, Munson EJ. 2011. Analysis of structural variability in and hydroxypropyl methylcellulose using a compaction simulator. Int
pharmaceutical excipients using solid-state NMR spectroscopy. AAPS J Pharm 348(1–2):46–53.
Pharm Sci Tech 12(3):821–833. 86. Yeh C, Altaf S, Hoag S. 1997. Theory of force transducer design op-
75. Cantor S, Hoag S, Ellison C, Khan M, Lyon R. 2011. NIR spec- timization for die wall stress measurement during tablet compaction:
troscopy applications in the development of a compacted multiparticu- Optimization and validation of split-web die using finite element anal-
late system for modified release. AAPS Pharm Sci Tech 12(1):262–278. ysis. Pharm Res 14(9):1161–1170.
76. Howland H, Hoag SW. 2013. Analysis of curing of a sustained re- 87. Heckel RW. 1961. Density–pressure relations in powder com-
lease coating formulation by application of NIR spectroscopy to monitor paction. Trans Am Inst Min Metall Petrol Eng 221:671–675.
changes physical-mechanical properties. Int J Pharm 452(1–2):82–91. 88. Kawakita K, Ludde HK. 1971. Some considerations on powder com-
77. Lim H, Dave VS, Kidder L, Lewis EN, Fahmy RM, Hoag SW. 2011. pression equations. Powder Technol 4:61–68.
Assessment of the critical factors affecting the porosity of roller com- 89. Haware RV, Bauer-Brandl A, Tho I. 2010. Comparative evalua-
pacted ribbons and the feasibility of using NIR chemical imaging to tion of the powder and compression properties of various grades and
evaluate the porosity distribution. Int J Pharm 410(1–2):1–8. brands of microcrystalline cellulose by multivariate methods. Pharm
78. Tabasi SH, Fahmy R, Bensley D, O’Brien C, Hoag SW. 2008. Quality Deve Tech 15(4):394–404.
by design, part I: Application of NIR spectroscopy to monitor tablet 90. Albers J, Knop K, Kleinebudde P. 2006. Brand-to-brand and batch-
manufacturing process. J Pharm Sci 97(9):4040–4051. to-batch uniformity of microcrystalline cellulose in direct tableting with
79. Tabasi SH, Fahmy R, Bensley D, O’Brien C, Hoag SW. 2008. Quality a pneumohydraulic tablet press. Pharm Ind 68(12):1420–1428.
by design, part III: Study of curing process of sustained release coated 91. Tho I, Bauer-Brandl A. 2011. Quality by design (QbD) approaches
products using NIR spectroscopy. J Pharm Sci 97(9):4067–4086. for the compression step of tableting. Expert Opin Drug Deliv
80. Tabasi SH, Fahmy R, Bensley D, O’Brien C, Hoag SW. 2008. Quality 8(12):1631–1644.
by design, part II: Application of NIR spectroscopy to monitor the coat- 92. Haware RV, Tho I, Bauer-Brandl A. 2009. Multivariate analysis
ing process for a pharmaceutical sustained release product. J Pharm of relationships between material properties, process parameters and
Sci 97(9):4052–4066. tablet tensile strength for "-lactose monohydrates. Eur J Pharm Bio-
81. Tabasi SH, Moolchandani V, Fahmy R, Hoag SW. 2009. Sustained pharm 73(3):424–431.
release dosage forms dissolution behavior prediction: A study of matrix 93. Haware RV, Shivagari R, Johnson PR, Staton S, Stagner WC,
tablets using NIR spectroscopy. Int J Pharm 382(1–2):1–6. Gupta MR. 2014. Application of multivariate methods to evaluate the
82. Gabrielsson J, Sjostrom M, Lindberg NO, Pihl AC, Lundstedt T. functionality of bovine- and vegetable-derived magnesium stearate. J
2006. Robustness testing of a tablet formulation using multivariate Pharm Sci 103(5):1466–1477.
design. Drug Dev Ind Pharm 32(3):297–307. 94. USFDA. 2004. Pharmaceutical cGMPs for the 21st cen-
83. Igne B, Shi Z, Drennen JK, 3rd, Anderson CA. 2014. Effects and tury: A risk-based approach http://www.fda.gov/downloads/Drugs/
detection of raw material variability on the performance of near- DevelopmentApprovalProcess/Manufacturing/QuestionsandAnswers
infrared calibration models for pharmaceutical products. J Pharm Sci onCurrentGoodManufacturingPracticescGMPforDrugs/UCM176374.
103(2):545–556. pdf.
84. Shah R, Tawakkul M, Khan M. 2008. Comparative evaluation of 95. Fu S, Thacker A, Sperger D, Boni R, Velankar S, Munson E, Block L.
flow for pharmaceutical powders and granules. AAPS Pharm Sci Tech 2010. Rheological evaluation of inter-grade and inter-batch variability
9(1):250–258. of sodium alginate. AAPS Pharm Sci Tech 11(4):1662–1674.

Dave et al., JOURNAL OF PHARMACEUTICAL SCIENCES DOI 10.1002/jps.24299

S-ar putea să vă placă și