You are on page 1of 10

Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10

Contents lists available at SciVerse ScienceDirect

Journal of Pharmaceutical and Biomedical Analysis


journal homepage: www.elsevier.com/locate/jpba

Review

Application of the near-infrared spectroscopy in the pharmaceutical technology


Marzena Jamrógiewicz ∗
Medical University of Gdansk, Faculty of Pharmacy, Department of Physical Chemistry, Hallera 107, 80-416 Gdansk, Poland

a r t i c l e i n f o a b s t r a c t

Article history: Near-infrared (NIR) spectroscopy is currently the fastest-growing and the most versatile analytical
Received 4 October 2011 method not only in the pharmaceutical sciences but also in the industry. This review focuses on recent NIR
Received in revised form 5 March 2012 applications in the pharmaceutical technology. This article covers monitoring, by NIR, of many manufac-
Accepted 7 March 2012
turing processes, such as granulation, mixing or drying, in order to determine the end-point of these
Available online 15 March 2012
processes. In this paper, apart from basic theoretical information concerning the NIR spectra, there
are included determinations of the quality and quantity of pharmaceutical compounds. Some exam-
Keywords:
ples of measurements and control of physicochemical parameters of the final medicinal products, such
Near-infrared spectroscopy
Pharmaceutical technology
as hardness, porosity, thickness size, compression strength, disintegration time and potential counter-
Quantitative feit are included. Biotechnology and plant drug analysis using NIR is also described. Moreover, some
Quality control disadvantages of this method are stressed and future perspectives are anticipated.
Multivariate analysis © 2012 Elsevier B.V. All rights reserved.
PAT

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
2. Recommendations for NIR application in the pharmaceutical sciences . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3. NIR in the pharmaceutical chemistry and physicochemistry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
4. Steps of pharmaceutical technology, controlled by NIR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
5. Plant drug analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
6. Biotechnological medicinal products tested by NIR spectroscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
7. Disadvantages of NIR spectroscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
8. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8

1. Introduction molecule causes particular bonds to vibrate in a manner which is


similar to that of a diatomic oscillator. Therefore, it is convenient to
Infrared spectral measurements have been used for a broad start from the diatomic molecule as the simplest vibrating system
range of applications – from analysis of liquids, gas compositions and then extend the concept to polyatomic molecules. Changes in
and solid substances to detailed characterization of each physi- light energy between the three regions of infrared lead to varying
cal state. One of the fundamental properties of chemical bonds is absorptions of different molecules and bonds, inducing different
that they exhibit vibrations at distinct frequencies. The vibrational types of vibrations. For instance, the least energetic far infrared
frequency of a chemical bond is intrinsic to the chemical bond of region of light (FIR) is absorbed by heavy atoms, such as some inor-
interest [1]. The electromagnetic energy of molecular vibration is ganic and organometallic substances, while the mid infrared area
defined as near-infrared (from 120 to 400 THz, from 2500 to 750 nm (MIR) is used for organic chemical analyses.
or from 12,500 to 4000 cm−1 ), infrared, or mid-infrared (from 30 Near-infrared (NIR) spectroscopy was not a popular technique
to 120 THz, from 2500 to 25,000 nm or from 4000 to 40 cm−1 ) and until 1960s. The current triumph of this method is owed to Karl
far-infrared between 300 GHz and 30 THz (that is from 25,000 nm Norris who recognized the potential of the use of NIR in industrial
to 1,000,000 nm). Infrared radiation absorbed by an individual practice for measurements of certain types of food, agricultural
components [2] and product quality control. This method gained
wide acceptance within the pharmaceutical industry, particularly
∗ Tel.: +48 58 349 16 56; fax: +48 58 349 16 52.
for the analysis of intact dosage forms and process monitoring [3],
and it could be applied for liquid, slurry, powdered or solid samples.
E-mail address: majam@gumed.edu.pl

0731-7085/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
doi:10.1016/j.jpba.2012.03.009
2 M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10

A potential molecule, intended to be analyzed by NIR spec-


troscopy, should possess the change of dipole moment and also a
large mechanical anharmonicity of the vibrating atoms [4]. There-
fore, a large variety of compounds is appropriate for analysis in the
range of near-infrared. Characteristic features of the NIR spectrum
are overtones and combination bands of CH, OH and NH function-
alities which dominate the spectrum, whereas the corresponding
overtones resulting from the fundamental absorption values of
the most intense MIR area are rarely represented [5]. Overtones
(electron excitations to higher energy levels) are similar to octaves
in a musical scale, like harmonics of the fundamental vibrational
frequencies [6]. Combination bands are the sum of two different
vibrations corresponding to different chemical bonds. Although
the absorbance values of combination bands and overtones are
smaller than transitions ones, in bulk materials (for example phar-
maceuticals) this feature is an advantage because of the material’s
properties, which allow obtaining high absorbance values.
Unfortunately, because of the overlapping of overtones and
combination of vibrations NIR spectra are much more complicated
for interpretation [7]. The total NIR spectrum contains up to four
overtones from the absorptions of methyl (C H), methylene (C H),
methoxy (C H), aromatic (C H), carbonyl associated (C H) groups,
N H from primary and secondary amides, N H from amines (pri-
mary, secondary, and tertiary), N H of amine salts, O H (alcohols
and water), S H, or C O groups [8]. In some papers clarification
of spectroscopic signals and wavenumber ranges characteristic for
each vibration–harmonics could be found [9].
Over the last few years near-infrared spectroscopy has devel-
oped into indispensable tool for academic research and industrial
quality control in a wide field of applications, from the synthetic
chemistry to the agriculture and from some life sciences to the Fig. 1. Critical stages of pharmaceutical manufacturing process of medicinal prod-
environmental analysis. It is the crucial method also in the pharma- ucts with the usage of NIR spectroscopy.
ceutical area, mainly in such branches as technology, microbiology,
toxicology, counterfeit detection, determination of physicochem-
spectroscopic studies were performed for a group of newly devel-
ical properties, stability testing and also quality control of a final
oped synthetic biopolymers. In this case, NIR spectra result mainly
product. Pharmaceutical technology needs NIR spectroscopy for
from these vibrational regions which could be divided into:
implementation of in- and on-line processes control in many phases
of manufacturing process (Fig. 1) [10–13].
- the first overtone of the hydroxyl stretching region;
The identification of specific wavelength regions is needed when
- the second overtone of the C H stretching mode, which could
changes in the signal obtained from the near-infrared spectrometer
be found in the high wavenumber region between 9000 and
indicate the changes which are proportional to the concentration of
6500 cm−1 ;
chemical components of pharmaceuticals, or represent the physi-
- the first overtone of the C H stretching vibrations, which could
cal characteristics of samples under analysis. Generally, qualitative
be found in the 5900–5350 cm−1 region;
and quantitative analysis performed by near-infrared spectroscopic
- the combination of O H stretching (mode) and second overtone
methods requires the application of multivariate calibration algo-
of the C O stretching mode, arising in the lower wavenumber
rithms (MCA), and, eventually, the usage of some chemometric
region between 5300 and 4800 cm−1 [17].
methods essential for model spectral response to chemical or phys-
ical properties of a calibration or learning model set. It is possible
and also quite recommended to create the spectral library of stan- The assignment of NIR absorption bands is important for simple
dard pharmaceutical materials using NIR spectroscopy as a rapid, band identification of active compound contained in a formula-
non-invasive technique for compound identification [14]. Very use- tion [18], as well as for the recognition of reaction product and
ful information about characteristic bands of active pharmaceutical also for the monitoring in the synthesis reactor. Petter et al. pre-
ingredients (APIs) or excipients is contained in recent publications sented a simple NIR spectroscopy method for monitoring of the
[15]. There is also a need for verification and identification of such peptide reaction between oxytocin and glutathione [19]. A qual-
simple compounds or residual solvents as methanol and ethanol in itative model of analysis allowed rapid identification of newly
pharmaceutical products. Two-dimensional (2D) correlation near- produced conjugates, while a quantitative method was aimed at
infrared spectroscopy sometimes is very useful for difficult and determination of the amount of oxytocin and glutathione degrada-
not typical analysis. This technique is useful for discovery of some tion products.
special vibration modes that cannot be clearly seen in the origi- A number of data and detailed spectra structure correlations
nal spectra. In example it is suited to distinguish between specific have been presented in a comprehensive handbook by Work-
bands for methanol and ethanol which are not clearly observed in man and may be helpful during interpretation of NIR spectra [10].
the combination vibration region of spectrum (5000–4500 cm−1 ) Author described the increased interest of spectroscopists, particu-
[16]. larly these involved in implementation of process of near-infrared
NIR spectroscopy is a valuable diagnostic tool which can be measurements, in this spectral region. Moreover, some examples
used for elucidation of comprehensive structural information of useful near-infrared absorption bands are presented in a book
of numerous biological samples. The comprehensive vibrational about NIR implementation by Katherine Bakeev [9].
M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10 3

2. Recommendations for NIR application in the radiation reaching the tablet core decreases and radiation reflected
pharmaceutical sciences by coating components increases [30,31].
The numerous possibilities of application of the NIR spec-
Advanced medicinal products require new generations of phar- troscopy and chemometric methods in the pharmaceutical area,
maceutical manufacture which must be ready for improved quality especially in the identification of raw materials, according to the
control of each step in the unit process of dosage form devel- quality control, as well as determination of moisture, are presented
opment [20]. In 2003, the Food and Drug Administration Agency precisely in the review article written by Luypaert et al. [32].
(FDA) announced Pharmaceutical Current Good Manufacturing The usage of NIR spectroscopy is very often based on a simple
Practices (cGMPs) for the 21st century in order to modernize all identification of active compound present in a drug. Civilization
the regulations and to get a better knowledge of the production problem with counterfeit medicinal products is a plague not only
processes. This document is recommended and respected all over in Asia [33]. Much harm is done to the customers because of tak-
the world. Generally, it is based on a risk approach of assurance ing up such drug products. The manufacturers suffer the greatest
of a pharmaceutical product quality and provides an implementa- loss because of counterfeit drug products and therefore they have
tion of Process Analytical Technology (PAT) in the process control started to develop various anti-counterfeiting practices such as dif-
of raw-materials, intermediate and final products [21,22]. Priority ferent analytical techniques, mainly NIR spectroscopy, thin layer
manners are focused on minimizing unwanted effects in the man- chromatography (TLC) or methods based on colorimetric detection
ufacturing process and on avoiding defects in the final products [34].
[23,24]. NIR spectroscopy allows selection of some characteristic spec-
The reason for increasing industrial application of NIR spec- tral bands and comparison of specific spectrum of original drug
troscopy in biomedical fields and pharmaceutical manufacture is product with unknown or doubtful one (e.g. with different phar-
the possibility of non-invasive mode of analysis [25]. It may be used maceutical excipients). For example, when the spectra of Chinese
in the determination of biochemicals in blood. Moreover, in spec- Sildenafil [35,36] and Viagra® are compared, the major variabil-
trophotometric methods used for process control it is crucial to ity between 5500 cm−1 and 5000 cm−1 could be observed [37].
receive signals from different points along the production line and As another example could serve “high-quality” Metronidazole,
to obtain the information from each measuring point. which counterfeit and original could be detected only by means
The implementation of the PAT system requires the identifica- of chemometric NIR data [38]. NIR spectroscopy was also success-
tion of critical parameters for each technological stage. Analytical fully applied during identification of confiscated drug blister, which
techniques chosen should allow the measurement of effects and proved to be bisoprolol-hemifumarate [39]. Important analyses of
statistical analysis in a very short time. Near-infrared spectroscopy counterfeit confirmation are frequently performed on the basis of
belongs to these techniques [26] and is suitable for on-, in- and NIR libraries created.
at-line process examinations. “In-line” means that process is mon- Special pharmaceutical excipients added to pharmaceutical
itored inside a reactor or a lyophilizer while “on-line” represents formulations create a unique composition which is difficult to
continuous control up to the end point (a sample flows through the identify by means of analytical methods recommended by Phar-
outer tube). Spectrum registration not directly at the production macopoeias [40–42] for particular drug compound, such as infrared
line, but close to it, is called “at-line”. spectroscopy or liquid chromatography. Excipients obtained from
An additional advantage of NIR is the possibility to handle and different manufacturers sometimes possess dissimilar physical
control a great number of industrial/technological variables which properties, particle size or purity. Therefore, there is a need of
must be optimized in manufacturing processes. In February 2009, view into the structure of each ingredient of the drug product
the European Medicines Agency (EMA) published the draft of a and of determination of its quality. For example, NIR spectroscopy
revised guideline on the use of NIR by the pharmaceutical indus- may be used for recognition and confirmation of some compounds
try and the data requirements for new submission and variations commonly used as tablet excipients, because in the process of
involving this technique [27]. According to cGMP the use of NIR manufacturing dosage forms all ingredients must be reliably iden-
spectroscopy is recommended in analytical procedure of techno- tified. In the current literature some examples about the capability
logical production of dosage forms. of FT-NIR spectroscopy to identify such substances as potassium
sorbate, sodium starch glycolate, calcium ascorbate, calcium car-
bonate, candelilla wax, maltosextrin, monohydrated or anhydrous
3. NIR in the pharmaceutical chemistry and lactose could be found [15]. The discrimination between anhydrous
physicochemistry and monohydrate form of lactose was done reliably inside USP vials.
Specific NIR band of lactose monohydrate corresponds to 1991 nm,
Near-infrared spectroscopy provides a rapid analysis of a wide while the anhydrous form possesses no band in this region. Apart
range of materials, giving a possibility of obtaining a large num- from complex mixtures analysis [43], all kinds of quantity [44–46]
ber of chemical and physicochemical parameters or even chemical and quality determination are performed in case of detection [47]
composition of some formulations as well. Various pharmaceuti- and location of chemical substances in a solid dosage forms [48].
cal parameters from tablet technology, such as hardness, particle The usage of NIR spectroscopy for comparison of manufacturing
size, compaction force, dissolution rate, or water content, can be series and batches of pharmaceutical products is fast and simple
quantitatively analyzed by NIR spectroscopy [3,28]. Good standard due to chemometrics [49].
of granulation step is acquired due to such quantity and quality In a study conducted by Bittner et al. [50], the simultaneous
tests as moisture content, particle size distribution and bulk den- identification and particle size determination of amoxicillin trihy-
sity [29]. Tablets made of fine-grained powder are often smooth drate in bulk materials was performed. NIR spectra of the observed
and easy to compress. The harder is a tablet, the smoother is its particle range of 7–22 ␮m showed an inverse relationship between
surface. Differences in properties cause reduction of light scat- absorbance (baseline offset) and particle size. Similar observation
tering and consequently build an upward shift in the spectral about the increase of baseline offset with decreasing particle size
baselines for smooth surface (small particles powders). Similar of measured material was also done by Abebe et al. [51].
situation is observed during determination of coating thickness The use of infrared spectroscopy is suggested by a Q6A
in NIR reflectance spectra. When the coating arises on a tablet, document of ICH as a way to perform and confirm the identi-
the absorbance values of core components decrease, because NIR fication of tablets and to obtain high-throughput and accurate
4 M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10

multiparametric data collection, as presented for a coated solid algorithm to global hyperspectral image [86]. NIR-CI system was
dosage form – thiamazole tablets containing 2, 4 and 8% of active also used for analysis of ibuprofen location in calcium phosphate
compound [52]. granules [87]. It was possible to investigate the chemical compo-
sition of some granules at high magnification and to gain useful
knowledge of the spatial location of the components.
4. Steps of pharmaceutical technology, controlled by NIR
None of the authors have investigated the influence of fluc-
tuations of environmental factors or time delay during spectra
There are many industrial or scientific reports about the usage of
recording on the images obtained. NIR-CI allows observation of
near-infrared spectroscopy for API determination not only directly
the contrast effect of the surface containing a number of ingredi-
in a formulation, but also as a raw material or during different man-
ents and therefore this method may be compared with microscope
ufacturing processes [10–13]. The latter depends on homogeneity
techniques.
of the formulation and compound’s stability. A non-destructive NIR
method of drug content prediction has recently been presented in
various aspects: 5. Plant drug analysis

- determination of API in a case of powders with different parti- In the last 40 years near-infrared spectroscopy became one of
cle size, which form agglomerates during blending. This research the most useful methods for analysis not only strictly pharmaceu-
described the problems during tablet manufacture, when the tical compounds but also proteins, dry matter of plant extracts or
presence of large agglomerates changes the content of API in carbohydrates quantity in many agriculture related products and
the direct compressive process and powder tableting [47]. The plant materials [3]. Some of them are intended to become medicinal
usage of NIR-Chemical Imaging Spectroscopy method allows to products. Recently, the use of NIR spectroscopy in determination
see the influence of particle size on the frequency of agglomerates of some minor compounds such as alkaloids, tocopherols, phenolic
formation; and glycoside compounds, glucosinolates, or essential oils in plant
- monitoring of unstable dosage forms such as suppositories con- materials has been reported [88,89]. In most cases authors claimed
taining acetylsalicylic acid [53], finding an alternative method for that the biggest challenge in the wider use of NIR spectroscopy
nimesulide analysis in pharmaceutical formulation, which is eas- is the interpretation of complex spectra obtained. Despite many
ier and faster in comparison with potentiometric titration [44]; multivariate analyses or chemometrics models used for the inter-
- quantification of low dosage pharmaceutical forms (for exam- pretation of spectra, the knowledge of the principles of molecular
ple syrup with acetaminophen, containing 16–24 mg/ml of the spectroscopy in natural products analysis is still the main barrier in
substance) which could be a very challenging task because of understanding the basis and functionality of the models developed.
relatively low sensibility of NIR spectroscopy methods [54], Pharmaceutical plants are often analyzed and classified due
development of a more suitable analytical method for oral solu- to the presence of some special marker compounds or medicinal
tions with dipyrone than iodometric titration [55]; compositions. Three species of Dendrobium (Orchidaceae), used for
- usage of appropriate NIR spectroscopy models for complex phar- stomach nourishing, promotion of production of body fluid and
maceutical matrixes, for example a hydroalcoholic gel containing fever reduction, were determined by NIR [90]. It was showed that
ketoprofen and two preservatives [56], examination of suitability the samples analyzed could be successfully verified on the basis
of a diffuse “transflection” technique for determination of diphen- of intensities of particular patterns of spectral bands at 5180 and
hydramine hydrochloride in pharmaceutical wafers with 2.5 cm 4900–4800 cm−1 . This study proved to be very useful and allows
diameter and a thickness of about 80 ␮m [57]. Samples during comprehensive evaluation of the different origins of Dendrobium,
measurements are placed directly on the round window, cover- as well as estimation of their clinical efficacy.
ing the integrating sphere, and masked with a diffusely reflecting Ginkgo biloba extract could be qualified as raw material intended
gold plate. NIR radiation after penetration through the sample is for use in pharmaceutical products. Some quality specifications
partially absorbed and partially reflected back into the sample, of G. biloba extract, intrinsic to its natural origin, show unpre-
allowing the radiation to penetrate through the wafer twice; dicted and uncontrolled variability, which has influence on the
- development of a method that allows the simultaneous iden- manufacturing process of solid dosage forms (viz. granulation and
tification and quantification of ranitidine in granulates for compression). Some of these variabilities could not be determined
compression, cores (as intermediate) and coated tablets [18]. by conventional quality control tests, therefore NIR method was
used for qualification of the batches of Ginkgo extract, according
to their different features, and for establishment of relationship
Japanese researches published some scientific reports about NIR
between some of the manufacturing steps. Several approaches
spectroscopic analyses of two APIs, coexisting in one tablet or form-
were evaluated, and the NIR method developed proved to be sen-
ing bilayer [58,59]. Due to irradiated NIR rays, penetrated through
sitive to changes in important quality specifications and therefore
the tablet, a method developed was non-destructive and effective.
adequate for qualification of incoming batches of G. biloba extract
The transmittance technique allows a simultaneous analysis of the
[91]. Nowadays, medicinal plant preparations are produced with
two layers. A highly sensitive method for determination of tablet
the highest quality control measurements, and each step of their
thickness fluctuation was also described [60].
manufacturing is also verified with PAT tools, such as NIR spec-
The utility of near-infrared spectroscopy for simultaneous in-
troscopy methods.
line drug and excipients quantification in manufacturing processes
is presented in many publications (Table 1).
During the last few years, growing interest in the use of 6. Biotechnological medicinal products tested by NIR
Near-Infrared Chemical Imaging (NIR-CI) method in the field of spectroscopy
pharmaceutical analysis is observed. The reason for increasing
popularity of this method is its ability to provide a wealth of Apart from applications in pharmaceutical technology, chem-
information from a single sample, including the quantitative com- istry or industry, near-infrared spectroscopy has also successfully
position analysis and distribution of ingredients. For example, been applied in the biotechnology field. Using near-infrared dyes,
the content and homogeneity distribution of acetylsalicylic acid it is possible to analyze DNA and protein molecules [92], to detect
in commercial tablets were analyzed by applying quantitative some genetically modified organisms by comparison of their DNA
Table 1
The utility of NIR spectroscopy in the pharmaceutical technology processes.

Analyzed compound Purpose Range [cm−1 ] Description Parameters Ref.

Polymorphism
Sulphathiazole Comparison with other methods for the analysis of the solid-state 10,000–4000 Quality and quantity Heating temp. 180 ◦ C; drying temp. 105 ◦ C Time of heating [61]
forms of sulfathiazole polymorphic forms, such as DSC, XPRD, NMR, 30 min.
Raman spectroscopy
Acetaminophen Polymorphic transformation monitored during a cooling 14,000–4000 Quality, compared Cooling from 60 ◦ C (1 ◦ C/min) [62]
crystallization process with crystallographic
data, XRD and DSC
Fluconazole Determination of pure crystalline fluconazole form II in a binary 12,500–4000 Samples obtained by A set of 12 samples covering a concentration range from [63]
polymorphic mixtures containing forms I and II comparing with XPRD mixing known 37.5 to 87.5% (37.5, 50, 62.5, 87.5% (w/w) was used to test
quantities of pure
polymorphic forms II
and III

M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10


Crystallization
Ibuprofen Comparative study of attenuated total reflectance (ATR), Fourier 12,000–4000 Quality and quantity Cooling (0.5 ◦ C/min) [64]
transform infrared (FTIR), and Fourier transform (FT) near-infrared
(NIR) spectroscopy for batch cooling crystallization of four different
crystallization systems
Cephalexin The crystallinity of cephalexin in microcrystalline cellulose was 8500–8300 Quality and quantity 6 types of standard material with various degrees of [65]
determined with the molecular interaction of a ground amorphous crystallinity were prepared by the physical mixing
solid investigation. X-ray powder diffraction profiles were
determinated
Trehalose The phase behavior of sugars in ice and lyophilized solids was 10,000–4000 Quality and quantity Amorphous trehalose was prepared using aqueous [66]
investigated for quantitate trehalose crystallization comparing with trehalose solutions frozen at −70 ◦ C and freeze-dried at
Raman spectroscopy −40 ◦ C with pressure of approximately 100 mbar for 72 h
Particle size
Dibasic calcium phosphate Quantification of the median particle size of dry powders 9100–4550 Quality and quantity Diameter of fractions: 53, 75, 106, 150, 180, 212, 250, [67]
300 ␮m time of mixing 10 min
Particle size distribution
Acetaminophen Quantitative determination of acetaminophen in granulates, 9000–4000 Quality and quantity Temp. 60 ◦ C, compacting pressure 195–215 Mpa, time [68]
establishment of the particle size distribution and examination of 180 min
average particle size was performed. The granulate samples were used
to obtain laboratory tablets examined in the study. Particle size
distribution was determined by using a PLS2 model allowing
correlation between the spectral data matrix and more than one
variable and prediction of various particle sizes with a single
calibration model
Humidity/water content
Unknown API In-line measurement of water content during the fluid bed drying 8500–4800 Quality and quantity Time: 30 min, temp. 60 ◦ C, air volume 1400–2300 m3 /h [69]
process of a drug product. This method is currently being applied on
the commercial scale to control the drying end point
Unknown API Determination of moisture content ranging from 1% to 8%. The 12,500–3600 Quantity Heating temp.: 105 ◦ C, time: 90 min [70]
calibration protocol covered 6 different moisture levels: 0.5%, 1%, 2%,
4%, 8% and 10%. The entire calibration was performed within 2 days.
Batches belonging to three different production campaigns were
chosen. Each sample was measured with the NIR spectrometer in
triplicate under two different temperature conditions: 5 and 25 ◦ C.
Lyophilized allergen The small amount of samples (10 mg/vial), despite of low moisture 14,300–4000 Quality – [71]
vaccine products content it was possible to use the NIR spectroscopy method
Blending
Acetaminophen Distribution of mixture components and simulated chemical 7140–6000 Quality and quantity Time: 0.5, 1, 2, 5, 10, 15, 20, 24, 30, 40 min; Rotation: [72]
concentrations of samples in a small scale with the usage of chemical 25 rpm
imaging
Acetaminophen Determination of blending end-point by the usage of a high rate of 11,000–6000 Variation of Time: 90 min, 200 spectra [73]
spectra collection (sensors positioned at the top and rotational axis of concentration
a bin-blender)

5
6
Table 1 (Continued)

Analyzed compound Purpose Range [cm−1 ] Description Parameters Ref.

Unknown API A real-time noninvasive NIR quantitative method was developed for 7400–6600; Quantity Rotation: 25 and 10 rpm; Time: 8, 20 min [74]
on-line blend uniformity analysis and transfer of the calibration model 6600–5500
from laboratory to industrial scale
Acetaminophen Monitoring of the concentration of acetaminophen blends during the 8800–6000 Quantity Rotation 1.000 rpm; Time: 2 min [75]
real time of the mixing process
Unknown API Based on a powder blend study, it was shown that the current practice 8300–4200 Quantity Rpm = 12; Time: 3, 5, 7, 9, 11, 13, 15, 17, 19, 21, 23, 25, 27, [76]
of calculating the % standard deviation for a given component within a 29, 60 min
specimen was not enough sensitive to assess blend uniformity
Tobramycin base Assessing blend uniformity in the context of the specific constraints of 10,000–4000 Quantity Rotation: 67 rpm; Time: 19 min [77]
complex seven-component blends and small batches
Tabletting
Micronized In-line quantification enabled the content analysis of individual tablets 10,200–5200 Quality and quantity 5 kg powder batch; Time: 30 min; Rotary tabletting [45]

M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10


chlorphenira-mine maleate in the production batch and detection of problems with content machine with 10 mm flat faced punches, operated at a
uniformity towards the end of the tabletting process. Furthermore, it speed of 10 rpm. Tablet weight: 530 mg
provided the assurance of in-process monitoring of content uniformity
of the particular excipients during the tabletting process
Compacts of lactose Monitoring of the physical quality of tablets, control of tableting 7100–4000 Quality and quantity Lactose monohydrate was blended with 0%, 0.25%, or 1.0% [78]
monohydrate with process and final ejection and production of density profiles by the MgSt for 30 s or 30 min. Compacts were prepared of each
lubricant usage of NIR-Chemical Imaging method blend, with compaction forces monitored by load cell
Compression
Metoprolol tartrate Demonstration of physical changes due to compression differences 7000–4000 Quality and quantity Tablets prepared by direct compression and wet [79]
(porosity, hardness) granulation. Time 10 s, pressure: 6.9–31 MPa
Production process of a Blend homogeneity, content uniformity, tablet coating thickness. A 12,500–5800 and Quality and quantity Duration of each mixing step: 25 min, rpm = 15, [31]
new potent drug total of 720 tablets from five batches were used as calibration set. 12,500–3600 compression forces 100–400 MPa
Target value was 2.98% (w/w). The validation set – 15 cores each
compacted at 100–400 MPa
Coating/film thickness
Sulfanilamide-based In-line control of the coating thickness (3% deviation from the actual 7500–4800 Quality and quantity Time: 160 min, number of tablets: 1.300, turnplate: [80]
tablets coated with HPMC, thickness), end-point determination for the fluid-bed coating process 200 rpm, fan flow: 125 m3 /h, temp. 238 ◦ C, humidity 40%,
PEG mixture spraying rate 1.1 mL/min, shaking bag interval: 60 s
MCC, lactose monohydrate, Correlation between in-line NIR spectra from a fluid-bed pellet coating 7500–5000 Quality and quantity Inlet air temp. 50 ◦ C, air flow: 60 m3 /h, spray rate of [81]
PVP, PEG 400, HPMC, process and two separate methods for determination of coating coating solution: 1 mL/min, atomization pressure 5 psi
fluorescein sodium salt thickness; laser diffraction particle size analysis and image analysis
MCC, lactose, PVP, starch Quantification of coat thickness for tablets coated at different process 9100–4000 Quality and quantity Tablets (6 mm diameter), 15% (w/w) solids coating [82]
conditions with the use of optical microscopy, micrometer, X-ray dispersion composed of 12% (w/w) hypromellose, 2% (w/w)
fluorescence, Raman and NIR spectroscopy. Tablet weight, thickness polyethylene glycol, and 1% (w/w) iron oxide in water
and diameter, hardness, surface roughness and color were determined
PE and 0.1% Aerosil 200 A novel small-scale coating system as a method of preparation a 9100–4500 Quality and quantity Tablets diameter between 7 and 11 mm, rotation speed [83]
calibration model used to predict the coating layer thickness. In-line 1 rpm, Six experiments with different rotations: 5, 10, 12,
coating analysis useful during the early stage of formulation studies 15, 20, 25 resulting in thickness from 25 to 400 ␮m
when the amount of drug is limited and formulations are being tested
on a small scale. Determination the coating thickness of tablets in a
drum coating process was performed. High-density polyethylene used
a strong NIR absorber to enable the detection of a core signal.
Porosity
Folic acid, microcrystalline Assessment of the effects of compaction variability in the tableting 11,520–6000 Quality and quantity Time of mixing: 12 min, tablet mass: 200.46 mg, diameter [84]
cellulose, crosscarmellose process on the performance of content prediction using NIR diffuse 10 mm, thickness 2.39 mm, resistance to crushing 95.33 N,
sodium, magnesium transmission mode. Examination of the effects of tablet thickness porosity 30.79%, dwell time 118.30 ms, upper compaction
stearate variation and/or porosity on the spectra. Elucidation of the force 3.90 kN
proportional contribution of various areas of a tablet to the final NIR
spectrum in diffuse transmission mode
Stability testing
Metoprolol tartrate tablets Presentation of differences in spectra obtained from metoprolol 7000–4000 Quality and quantity 1. Temp. 25 ◦ C, humidity: 60% [85]
tartrate tablets packed in the original high density polyethylene 2. Temp. 40 ◦ C, humidity 75%; Time: 13 and 52 weeks
containers or in USP Class A unit-dose blister packs
M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10 7

fragments [93], and above all, to examine the activities and roles of a processes as well as provides a way of qualification. Such quantita-
particular gene expression in some seed phenotypes with regard to tive studies are very important also in toxicological measurements
their physiochemical composition [94]. Near-infrared spectroscopy of wastes from pharmaceutical manufactures.
characteristics have also been used for localization of some oxida- During work with samples which may suffer any kind of peri-
tive DNA damages [95]. odic or seasonal change, NIR operators should be aware of any
The development of some new analytical tools, including spec- changes in the reference method which may negatively impact the
troscopic sensors and data analysis, still continues. Miniaturized calibration process. Keeping the sample calibration pool updated
tools for remote environmental monitoring, based on integrated by adding new samples when required and using standards for
circuit technology, have recently been developed, leading to the periodic examinations should be an obligatory practice by NIR cal-
marriage of biosensors and the lab-on-a-chip [23]. Near-infrared ibrations, which means that standardization tasks are periodically
spectroscopy has many applications as a process monitoring and unavoidable [101].
process supervision technique, also in the context of bioman- Scientific and academic groups of pharmacists and chemists are
ufacturing. An industrial pilot-plant process of mammalian cell not especially involved in the applications of NIR method. This may
cultivation presented the use of on-line in situ NIR application by be due to the fact that during academic work smaller amounts of
means of an immersion transflectance [96]. For animal cell culture samples are prepared in comparison with industrial conditions.
processes, important recommendations for improving efficiency Routine in work with reference methods is accepted and suffi-
and for ensuring the quality of the final product were defined cient when a great number of samples are analyzed. Therefore, NIR
by PAT. An analytical procedure, using FT-NIR spectroscopy and spectroscopy is the most popular method used by people working
chemometrics with multivariate techniques, was developed for or connected with industry, where PAT, FDA or Pharmacopoeias
the determination of the fruit authenticity and for the quantifica- recommendations play a primary role.
tion of the fruit content [97]. Spectral information was correlated A proper interpretation of results obtained after application
with the fruit content and gravimetric data obtained from sequen- of NIR spectroscopy in quality as well as quantity determina-
tial fractionation of the alcohol-insoluble residue. The contents tion, which usually are numerous, chemometric data, is a difficult
of rhamnose, fucose, arabinose, xylose, mannose, galactose, and task. A person who operates NIR device must be well prepared to
glucose in the hemicellulose fraction of apricots, peaches, and choose the best from many spectra obtained by pre-treatments
pumpkins were correlated and determined. steps, calibration and validation processes for further calculation.
NIR is also an improved method for evaluation of the amount Apart from software developed for a method planning and setting
of physiologically acceptable polymer moieties (PEG) conjugated for special analysis (establishment of API concentration, end-point
to a protein. None of the existing analytical methods (colorimetry, determination or identification) NIR operator needs a harder view
HPLC, phase-partitioning) are reliable enough for the quantitative to manipulate spectra obtained in order to create good fitted
determination of the amount of polymer molecules bound to a pro- mathematic results. Good analytical practice requires collabora-
tein. This patented method [98] involves contact of the sample with tion between analyst, chemist, technologist and also well-educated
a light source in the near-infrared spectrum and then measurement mathematician or statistician for proper calculation of all data.
of relative absorbance levels. Inventors of the method claimed to NIR spectroscopy provides an alternative for validated con-
determine over a range of the near-infrared wavelengths the num- ventional methods. Although, it is important to keep in mind
ber of polymer molecules, conjugated to the protein or protein restrictions about sensitivity and equipment or environmental
complex. The essence and important advantage of the near-infrared specificity of this method. It cannot be repeated easily in official
spectroscopy is that this technique is useful in studies in which control laboratories, so the reference method needs to remain the
other methods are inadequate or ineffective. important part of the official specifications. Current software allows
A large number of biomolecules with potential pharmaceutical development of NIR spectroscopy methods with minimal under-
application are produced just as all medicinal products on the basis standing of the relevant chemometrics, which creates a high risk
of Quality-by-Design (QbD) initiative [99]. Desired biological prop- of obtaining invalid results due to some unknown hidden variables
erties of the molecules are achieved during development of such [102].
factors as serum half-life, formulation, aggregation propensity, and Choice of proper parameters during pre-treatments operations
immunogenicity, which are important for the production of the or spectral pre-processing methods, which are needed during
biologically active molecule. the near-infrared spectroscopy application, is regulated by a set
of mathematical procedures before development of a calibration
model. Mathematical pre-treatment of spectrum reduces noise
7. Disadvantages of NIR spectroscopy or background information (smoothing techniques) and increases
signal from the chemical compound analyzed (differentiation pro-
Near-infrared spectroscopy possesses several advantages over cess). Any pre-treatment technique should succeed in a robust
other analytical techniques used in the pharmaceutical technology model with good predictive ability. In general, pre-processing
but there are also some disadvantages of this method (Table 2). methods can be defined as baseline correction–normalization,
One of the main disadvantages of NIR spectroscopy used in phar- enhancement, and statistical filtering of the signal obtained. The
maceutical technology is a problem with aqueous environment effects of several pre-processing methods used on absorbance spec-
(solutions of medicinal products or active pharmaceutical ingre- tra obtained by reflectance mode instrument are presented by
dients alone) for direct quantity determinations. High absorbance Agelet and Hurburgh [8]. The offset baseline correction could be
of water, which leads to a broad peak on a spectrum, creates a limit observed when pre-processing method with SNV (standard nor-
of detection for other compounds that are present in the sample mal variation) is used. With the usage of MSC (multiple scatter
analyzed in smaller quantities. In situ NIR spectroscopy technique correction) and first and second Savitzky–Golay derivatives pre-
allows monitoring an activated sludge reactor by estimation of the processing methods, after elimination of scatter effects, the spectra
nitrate and oxygen concentration (N NO3 − ) and the amount of obtained are more grouped together. In each case, the NIR operator
total suspended solids [100]. Characteristic spectra were collected is forced to choose a proper way to achieve satisfactory results. This
with the use of immersion probes on the settler. This alternative step of analysis may cause some difficulties.
method, both with UV–Vis spectroscopy, presents clear advan- According to Lambert–Beer’s law, in the case of calibration of
tages during monitoring and controlling of wastewater treatment spectroscopy method, the linear correlation between analyte or
8 M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10

Table 2
Several advantages and disadvantages of NIR spectroscopy application in the pharmaceutical technology and manufacturing.

Advantages Disadvantages

Non-destructive and non-invasive method Low sensitivity of the signal limit by the determination of substances with
concentration below 0.1% (w/w)
Rapid spectral measurements (<1 min) The initial high financial investment for the instrumentation
Low-cost of analysis – no chemical reagents needed, single operator can analyze a Development of calibration models requires high trained personnel
large number of samples
A possibility of scanning several spectra on the same object permits obtaining Accurate and robust calibration requires a large data set incorporated with
more representative sample composition and more accurate result of analysis large variation, which is often difficult to obtain
Sample requires minimal (drying and grinding) or no preparation Requires a continuous maintenance of the calibration data set
Several constituents of the same sample can be measured at the same time It is difficult to transfer calibration between instruments of the same
manufacture or between different manufactures
Easily applicable in different environments such as industry or laboratory It requires an accurate chemical and physical analysis of reference samples
Measurements can be carried out on/in/at line
The possibility of using optical probes allows analysis of the sample in situ
The availability of portable instruments permits obtaining spectra directly in the
field, which is useful for following some active processes
Ability to penetrate glass containers
Can also be used for particulate level (e.g., particle size) and bulk level (e.g., water
content) measurements with proper calibration

property measured and its absorbance should be the first assump- References
tion. NIR calibration models correlate either raw or pre-processed
spectra with one or more chemical-physical properties of a set of [1] P. Atkins, Physical Chemistry, fifth ed., Freeman, New York, 1994.
[2] J.R. Hart, K.H. Norris, C. Golumbic, Determination of the moisture content of
samples. As complicated as it may sound, there are several well- seeds by near-infrared spectrophotometry of their methanol extracts, Cereal
developed calibration methods which are proved to work with Chem. 39 (1962) 94–99.
most of NIR applications and also are included in all chemometric [3] C.A. Anderson, J.K. Drennen, E.W. Ciurczak, Pharmaceutical applications of
near-infrared spectroscopy, in: D.A. Burns, E.W. Ciurczak (Eds.), Handbook of
software packages. Near-Infrared Analysis, Taylor & Francis Group, 2008, pp. 585–612.
Unfortunately, NIR spectroscopy has received some bad public- [4] M.J. Pelletier, C.C. Pelletier, Spectroscopic theory for chemical imaging, in: S.
ity as a “black box” technique. Something is introduced into the Sasic, Y. Ozaki (Eds.), Raman, Infrared, and Near Infrared Chemical Imaging,
Wiley, 2010, pp. 1–20.
black box and something out of this box is replaced without the [5] H.W. Siesler, Y. Ozaki, S. Kawata, H.M. Heine, Near-infrared Spectroscopy
knowledge of a mechanism or detailed calculation. Testing of a Principles, Instruments, Applications, fourth ed., Wiley, Weinhein, 2002.
method is combined with verification and validation, while veri- [6] F.H. Long, Vibrational spectroscopic methods for quantitative analysis, in: K.
Huynh-Ba (Ed.), Handbook of Stability Testing in Pharmaceutical Develop-
fication means checking the components of the system in terms of
ment, Springer Science, New York, 2009, pp. 224–228.
their compliance with the attached specifications. Moreover, it is [7] E. Alm, R. Bro, S.B. Engelsen, B. Karlberg, R.J.O. Torgrip, Vibrational overtone
often said that chemometric correlation requires a lot of faith in combination spectroscopy (VOCSY) – a new way of using IR and NIR data,
numbers that many spectroscopists do not possess [103]. Making a Anal. Bioanal. Chem. 388 (2007) 179–188.
[8] L.E. Agelet, C.R. Hurburgh, A tutorial on near-infrared spectroscopy and its
lot of analyses on a modern instrument without advanced courses calibration, Crit. Rev. Anal. Chem. 40 (2010) 246–260.
about proper usage of complicated software may be in fact a totally [9] M.B. Simpson, Near-infrared spectroscopy for process analytical technology:
bad experience. theory, technology and implementation, in: K.A. Bakeev (Ed.), Process Analyt-
ical Technology. Spectroscopic Tools and Implementation Strategies for the
NIR spectroscopy has increasingly been adopted as an analytical Chemical and Pharmaceutical Industries, John Wiley & Sons, New York, 2010,
tool (PAT) in pharmaceutical industry, mainly on the step of pre- pp. 40–51.
formulation and quality control of manufacturing process. The fast [10] J. Workman Jr., A review of process near infrared spectroscopy: 1980–1994,
J.Near Infrared Spectrosc. 1 (1993) 221–245.
and harmless analysis performed and samples characterization as [11] G. Reich, Near-infrared spectroscopy and imaging: basic principles and phar-
well as this method flexibility is the main reasons for popularity maceutical applications, Adv. Drug Del. Rev. 57 (2005) 1109–1143.
and particular usage of NIR. [12] M. Blanco, J. Coello, H. Iturriaga, S. Maspoch, C. de la Pezuela, Near-infrared
spectroscopy in the pharmaceutical industry, Analyst 123 (1998) 135R–150R.
[13] T. De Beer, A. Burggraeve, M. Fonteyne, L. Searens, J.P. Remon, C. Vervaet, Near
8. Conclusions infrared and Raman spectroscopy for the in-process monitoring of pharma-
ceutical production processes, Int. J. Pharm. 417 (2010) 32–47.
[14] J. Workman, L. Weyer, Practical Guide to Interpretive Near Infrared, Taylor
There are many applications of near-infrared spectroscopy in and Francis, London, New York, 2008, pp. 117–286.
the pharmaceutical technology. In addition, many difficult techno- [15] H.R.H. Ali, H.G.M. Edwards, I.J. Scowen, Noninvasive in situ identification and
logical problems in the scientific or industrial field may be solved band assignments of some pharmaceutical excipients inside USP vials with
FT-near-infrared spectroscopy, Spectrochim. Acta A: Mol. Biomol. Spectrosc.
due to this popular PAT tool. NIR spectra band depends on the phys-
72 (2009) 890–896.
ical or chemical nature of the sample and allows its identification [16] I. Noda, Progress in two-dimensional (2D) correlation spectroscopy, J. Mol.
as well as determination of physical or chemical parameters of the Struct. 799 (2006) 2–15.
[17] R.K. Bista, R.F. Bruch, A.M. Covington, Vibrational spectroscopic studies of
sample. The advantages of NIR spectroscopy method are numer-
newly developed synthetic biopolymers, Biopolymers 93 (2010) 403–417.
ous: non-invasive and non-destructive techniques, no preparation [18] S.S. Rosa, P.A. Barata, J.M. Martins, J.C. Menezes, Development and valida-
of sample, high frequency of spectrum acquisition, as well as large tion of a method for active drug identification and content determination of
number of molecules which could be quantified. NIR method is ranitidine in pharmaceutical products using near-infrared reflectance spec-
troscopy: a parametric release approach, Talanta 75 (2008) 725–733.
crucial not only in scientific, but also in practical sense. It requires [19] C.H. Petter, N. Heigl, S. Bachmann, V.A.C. Huck-Pezzei, M. Najam-ul-Haq,
cooperation between good chemists, pharmaceutical technologists R. Bakry, A. Bernkop-Schnürch, G.K. Bonn, C.W. Huck, Near infrared spec-
and mathematics for chemometric calculations and results descrip- troscopy compared to liquid chromatography coupled to mass spectrometry
and capillary electrophoresis as a detection tool for peptide reaction moni-
tion, and this trend corresponds to the interdisciplinary of science toring, Amino Acids 34 (2008) 605–616.
also in other fields of manufacturing practice. NIR spectroscopy will [20] K.R. Morris, S.L. Nail, G.E. Peck, S.R. Byrn, U.J. Griesser, J.G. Stowell, S.-J. Hwang,
certainly play a key role in the analysis of many industrial pro- K. Park, Advances in pharmaceutical materials and processing, PSTT 1 (1998)
235–245.
cesses after implementation this method into the real-time process [21] M.C. Sarraguca, J.A. Lopes, Quality control of pharmaceuticals with NIR: From
monitoring during continuous pharmaceutical production. lab to process line, Vibr. Spectrosc. 49 (2009) 204–210.
M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10 9

[22] FDA. Guidance for industry: PAT – a framework for innovative pharmaceutical [52] L. Alvarenga, D. Ferreira, D. Altekruse, J.C. Menezes, D. Lochmann, Tablet iden-
development. Manufacturing, and quality assurance, Pharmaceutical CGMPs, tification using near-infrared spectroscopy (NIRS) for pharmaceutical quality
2004. control, J. Pharm. Biomed. Anal. 48 (2008) 62–69.
[23] M. Blanco, M. Alcala, Use of near-infrared spectroscopy for off-line mea- [53] E. Otsuka, H. Abe, M. Aburada, M. Otsuka, A nondestructive prediction of the
surements in the pharmaceutical industry, in: K.A. Bakeev (Ed.), Process drug content of an aspirin suppository by near-infrared spectroscopy, Drug
Analytical Technology, Spectroscopic Tools and Implementation, Strategies Dev. Ind. Pharm. 36 (2010) 839–844.
for the Chemical and Pharmaceutical Industries, Blackwell Publishing, New [54] E. Ziémons, J. Mantanus, P. Lebrun, E. Rozet, B. Evrard, Ph Hubert,
York, 2005, pp. 13–20, 362–376. Acetaminophen determination in low-dose pharmaceutical syrup by NIR
[24] R. Jachowicz, K. Woyna-Orlewicz, PAT – process analytical technology as a spectroscopy, J. Pharm. Biomed. Anal. 53 (2010) 510–516.
manufacturing process optimization strategy, Farm. Pol. 66 (2010) 209–214. [55] M.H. Ferreira, J.F.F. Gomes, M.M. Sena, Development and validation of
[25] M. Blanco, I. Villarroya, NIR spectroscopy: a rapid-response analytical tool, multivariate calibration model for determination of dipyrone in oral
Trends Anal. Chem. 21 (2002) 240–250. solutions by near infrared spectroscopy, J. Braz. Chem. Soc. 20 (2009)
[26] J. Märk, M. Andre, M. Karner, ChW. Huck, Prospects for multivariate classifi- 1680–1686.
cation of a pharmaceutical intermediate with near-infrared spectroscopy as [56] M. Blanco, M. Alcala, M. Bautista, Pharmaceutical gel analysis by NIR spec-
a process analytical technology (PAT) production control supplement, Eur. J. troscopy: Determination of the active principle and low concentration of
Pharm. Biopharm. 76 (2010) 320–327. preservatives, Eur. J. Pharm. Sci. 33 (2008) 409–414.
[27] EMEA, Guideline on the use of near-infrared spectroscopy by the pharmaceu- [57] M. Haag, M. Brüning, K. Molt, Quantitative analysis of diphenhydramine
tical industry and the data requirements for new submissions and variations, hydrochloride in pharmaceutical wafers using near infrared and Raman spec-
European Medicines Agency, London, 16 (2009) 1–30. troscopy, Anal. Bioanal. Chem. 395 (2009) 1777–1785.
[28] M.P. Freitas, A. Sabadin, L.M. Silva, F.M. Gianotti, D.A. do Couto, E. Tonhi, R.S. [58] Y. Dou, N. Qu, B. Wang, Y.Z. Chi, Y.L. Ren, Simultaneous determination of two
Medeiros, G.L. Coco, V.F.T. Russo, Prediction of drug dissolution profiles from active components in compound aspirin tablets using principal component
tablets using NIR diffuse reflectance spectroscopy: a rapid and nondestructive artificial neural networks (PC-ANNs) on NIR spectroscopy, Eur. J. Pharm. Sci.
method, J. Pharm. Biomed. Anal. 39 (2005) 17–21. 32 (2007) 193–199.
[29] M. Alcala, M. Blanco, M. Bautista, J.M. Gonzalez, On-line monitoring of a gran- [59] M. Ito, T. Suzuki, S. Yada, H. Nakagami, H. Teramoto, E. Yonemochia, K. Terada,
ulation process by NIR spectroscopy, J. Pharm. Sci. 99 (2010) 336–345. Development of a method for nondestructive NIR transmittance spectro-
[30] J.D. Kirsch, J.K. Drennen, Determination of film-coated tablet parameters by scopic analysis of acetaminophen and caffeine anhydrate in intact bilayer
near-infrared spectroscopy, J. Pharm. Biomed. Anal. 13 (1995) 1273–1281. tablets, J. Pharm. Biomed. Anal. 53 (2010) 396–402.
[31] J.J. Moes, M.M. Ruijken, E. Gout, H.W. Frijlink, M.I. Ugwoke, Application [60] M. Ito, T. Suzuki, S. Yada, A. Kusai, H. Nakagami, E. Yonemochi, K. Terada,
of process analytical technology in tablet process development using NIR Development of a method for the determination of caffeine anhydrate in
spectroscopy: Blend uniformity, content uniformity and coating thickness various designed intact tablets by near-infrared spectroscopy: a comparison
measurements, Int. J. Pharm. 357 (2008) 108–118. between reflectance and transmittance technique, J. Pharm. Biomed. Anal. 47
[32] J. Luypaert, D.L. Massart, Y.V. Heyden, Near-infrared spectroscopy applica- (2008) 819–827.
tions in pharmaceutical analysis, Talanta 72 (2007) 865–883. [61] Y. Hu, A. Erxleben, A.G. Ryder, P. McArdle, Quantitative analysis of sulphathia-
[33] Y.C. Feng, X.B. Zhang, C.Q. Hu, Construction of identification system for non- zole polymorphs in ternary mixtures by attenuated total reflectance infrared,
invasive analysis of macrolides tablets using near infrared diffuse reflectance near-infrared and Raman spectroscopy, J. Pharm. Biomed. Anal. 53 (2010)
spectroscopy, J. Pharm. Biomed. Anal. 51 (2010) 12–17. 412–420.
[34] K.T. Ram, P.S. Saumil, Y. Agrawal, Pharmaceutical counterfeiting and analyti- [62] I.-Ch. Wang, M.-J. Lee, D.-Y. Seo, H.-E. Lee, Y. Choi, W.-S. Kim, Ch-S. Kim,
cal authentication, Curr. Pharm. Anal. 7 (2011) 54–61. M.-Y. Jeong, G.J. Choi, Polymorph transformation in paracetamol monitored
[35] T. Liu, Y.C. Feng, D.Q. Song, C.Q. Hu, Selection of characteristic spectral bands by in-line NIR spectroscopy during a cooling crystallization process, AAPS
for the analysis by the NIR correlation coefficient method, J. Chin. Pharm. Sci. PharmSciTech. 12 (2011) 764–770.
20 (2011) 83–91. [63] E. Ziemons, H. Bourichi, J. Mantanus, E. Rozet, P. Lebrun, E. Essassi, Y. Cher-
[36] B.J. Venhuis, G. Zomer, M.J. Vredenbregt, D. de Kaste, The identification of (−)- rah, A. Bouklouze, Ph Hubert, Determination of binary polymorphic mixtures
trans-tadalafil tadalafil, and sildenafil in counterfeit Cialis® and the optical of fluconazole using near infrared spectroscopy and X-ray powder diffrac-
purity of tadalafil stereoisomers, J. Pharm. Biomed. Anal. 51 (2010) 723–727. tion: a comparative study based on the pre-validation stage results, J. Pharm.
[37] P.Y. Sacre, E. Deconinck, T. De Beer, P. Courselle, R. Vancauwenberghe, P. Chiap, Biomed. Anal. 55 (2011) 1208–1212.
J. Crommen, J.O. De Beer, Comparison and combination of spectroscopic tech- [64] S.S. Kadam, E. van der Windt, P.J. Daudey, H.J.M. Kramer, A comparative study
niques for the detection of counterfeit medicines, J. Pharm. Biomed. Anal. 53 of ATR–FTIR and FT-NIR spectroscopy for in situ concentration monitoring
(2010) 445–453. during batch cooling crystallization processes, Cryst. Growth Des. 10 (2010)
[38] O.Y. Rodionova, A.L. Pomerantsev, NIR based approach to counterfeit-drug 2629–2640.
detection, Trends Anal. Chem. 29 (2010) 781–938. [65] Y. Fukui, M. Otsuka, Determination of the crystallinity of cephalexin in phar-
[39] T. Puchert, D. Lochmann, J.C. Menezes, G. Reich, Near-infrared chemical imag- maceutical formulations by chemometrical near-infrared spectroscopy, Drug
ing for counterfeit drug identification, J. Pharm. Biomed. Anal. 51 (2010) Develop. Ind. Pharm. 36 (2010) 72–80.
138–145. [66] B. Connolly, T.W. Patapoff, Y.J. Wang, J.M. Moore, T.J. Kamerzell, Vibrational
[40] European Pharmacopoeia 6.0, 2007. spectroscopy and chemometrics to characterize and quantitate trehalose
[41] United States Pharmacopoeia, 2009. crystallization, Anal. Biochem. 399 (2010) 48–57.
[42] British Pharmacopoeia, 2007. [67] D.R. Ely, M. Thommes, M.T. Carvajal, Analysis of the effects of particle size and
[43] D. Xiang, M. Konigsberger, B. Wabuyele, K. Hornung, J. Cheney, Development densification on NIR spectra, Colloids Surf. A: Physicochem. Eng. Aspects 331
of robust quantitative methods by near-infrared spectroscopy for rapid phar- (2008) 63–67.
maceutical determination of content uniformity in complex tablet matrix, [68] M. Blanco, R. Cueva-Mestanza, A. Peguero, Controlling individual steps in
Analyst 134 (2009) 1405–1415. the production process of paracetamol tablets by use of NIR spectroscopy,
[44] W.F.C. Rocha, A.L. Rosa, J.A. Martins, R.J. Poppi, Determination and validation J. Pharm. Biomed. Anal. 51 (2010) 797–804.
of nimesulide in pharmaceutical formulation by near infrared spectroscopy, [69] A. Peinado, J. Hammond, A. Scott, Development, validation and transfer
J. Braz. Chem. Soc. 21 (2010) 1929–1936. of a Near Infrared method to determine in-line the end point of a flu-
[45] A.D. Karande, P.W.S. Heng, C.V. Liew, In-line quantification of micronized idized drying process for commercial production batches of an approved
drug and excipients in tablets by near infrared (NIR) spectroscopy. Real time oral solid dose pharmaceutical product, J. Pharm. Biomed. Anal. 54 (2010)
monitoring of tabletting process, Int. J. Pharm. 396 (2010) 63–74. 13–20.
[46] C.V. Liew, A.D. Karande, P.W.S. Heng, In line quantification of drug and excip- [70] J. Mantanus, E. Ziemons, P. Lebrun, E. Rozet, R. Klinkenberg, B. Streel, B. Evrard,
ients in cohesive powder blends by near infrared spectroscopy, Int. J. Pharm. Ph Hubert, Moisture content determination of pharmaceutical pellets by near
386 (2010) 138–148. infrared spectroscopy: method development and validation, Anal. Chim. Acta
[47] J. Mantanus, E. Ziémons, P. Lebrun, E. Rozet, R. Klinkenberg, B. Streel, B. 642 (2009) 186–192.
Evrar, Ph Hubert, Active content determination of non-coated pharmaceu- [71] Y. Zheng, X. Lai, S.W. Bruun, H. Ipsen, J. Nedergaard, H. Løwenstein, I.
tical pellets by near infrared spectroscopy: method development, validation Søndergaard, S. Jacobsen, Determination of moisture content of lyophilized
and reliability evaluation, Talanta 80 (2010) 1750–1757. allergen vaccines by NIR spectroscopy, J. Pharm. Biomed. Anal. 46 (2008)
[48] W. Li, A. Woldu, R. Kelly, J. McCool, R. Bruce, H. Rasmussen, J. Cunningham, D. 592–596.
Winstead, Measurement of drug agglomerates in powder blending simulation [72] H. Ma, C.A. Anderson, Characterization of pharmaceutical powder blends by
samples by near infrared chemical imaging, Int. J. Pharm. 350 (2008) 369–373. NIR chemical imaging, J. Pharm. Sci. 97 (2008) 3305–3320.
[49] Y. Roggo, P. Chalus, L. Maurer, C. Lema-Martinez, A. Edmond, N. Jent, A review [73] Z. Shi, R.P. Cogdill, S.M. Short, C.A. Anderson, Process characterization of pow-
of near infrared spectroscopy and chemometrics in pharmaceutical technolo- der blending by near-infrared spectroscopy. Blend end-points and beyond, J.
gies, J. Pharm. Biomed. Anal. 44 (2007) 683–700. Pharm. Biomed. Anal. 47 (2008) 738–745.
[50] L.K.H. Bittner, N. Heigl, C.H. Petter, M.F. Noisternig, U.J. Griesser, G.K. Bonn, [74] Y. Sulub, B. Wabuyele, P. Gargiulo, J. Pazdan, J. Cheney, J. Berry, A. Gupta, R.
C.W. Huck, Near-infrared reflection spectroscopy (NIRS) as a successful tool Shah, H. Wu, M. Khan, Real-time on-line blend uniformity monitoring using
for simultaneous identification and particle size determination of amoxicillin near-infrared reflectance spectrometry: a noninvasive off-line calibration
trihydrate, J. Pharm. Biomed. Anal. 54 (2011) 1059–1064. approach, J. Pharm. Biomed. Anal. 49 (2009) 48–54.
[51] S.B. Abebe, X.Z. Wang, R. Li, K.J. Roberts, X. Lai, The information content in [75] A.U. Vanarase, M. Alcalà, J.I. Jerez Rozo, F.J. Muzzio, R.J. Romañach, Real-time
NIR spectral data for slurries of organic crystals, Powder Technol. 179 (2008) monitoring of drug concentration in a continuous powder mixing process
176–183. using NIR spectroscopy, Chem. Eng. Sci. 65 (2010) 5728–5733.
10 M. Jamrógiewicz / Journal of Pharmaceutical and Biomedical Analysis 66 (2012) 1–10

[76] M. Römer, J. Heinämäki, C. Strachan, N. Sandler, J. Yliruusi, Prediction of tablet [90] C. Wang, B. Xiang, W. Hang, Application of two-dimentional near-infrared
film-coating thickness using a rotating plate coating system and NIR spec- (2D-NIR) correlation spectroscopy to the discrimination of three species of
troscopy, AAPS PharmSciTech. 9 (2008) 1047–1053. Dendrobium, J. Chemometrics 23 (2009) 463–470.
[77] T. Puchert, D. Lochmann, J.C. Menezes, G. Reich, A multivariate approach for [91] S.S. Rosa, P.A. Barata, J.M. Martins, J.C. Menezes, Near-infrared reflectance
the statistical evaluation of near-infrared chemical images using symmetry spectroscopy as a process analytical technology tool in Ginkgo biloba extract
parameter image analysis (SPIA), Eur. J. Pharm. Biopharm. 78 (2011) 117–124. qualification, J. Pharm. Biomed. Anal. 47 (2008) 320–327.
[78] I. Storme-Paris, I. Clarot, S. Esposito, J.C. Chaumeil, A. Nicolas, F. Brion, A. [92] F.-F. Fu, R. Ye, S.-P. Xu, H.-W. Xue, Studies on rice seed quality through analysis
Rieutord, P. Chaminade, Near infrared spectroscopy homogeneity evaluation of a largescale T-DNA insertion population, Cell Res. 19 (2009) 380–391.
of complex powder blends in a small-scale pharmaceutical preformulation [93] E. Michelini, P. Simoni, P. Cevenini, L. Mezzanotte, A. Roda, New trends in
process, a real-life application, Eur. J. Pharm. Biopharm. 72 (2009) 189–198. bioanalytical tools for the detection of genetically modified organisms: an
[79] C.D. Ellison, B.J. Ennis, M.L. Hamad, R.C. Lyon, Measuring the distribution of update, Anal. Bioanal. Chem. 392 (2008) 355–367.
density and tabletting force in pharmaceutical tablets by chemical imaging, [94] L. Munck, B.M. Jespersen, From discovery of high lysine barley endosperm
J. Pharm. Biomed. Anal. 48 (2008) 1–7. mutants in the 1960–70s to new holistic spectral models of the phenome
[80] R.B. Shah, M.A. Tawakkul, M.A. Khan, Process analytical technology: chemo- and of pleiotropy in 2008, in: Q.Y. Shu (Ed.) Induced Plant Mutations in the
metric analysis of Raman and near infrared spectroscopic data for predicting Genomics Era, Rome, 2009, pp. 419–422.
physical properties of extended release matrix tablets, J. Pharm. Sci. 96 (2007) [95] A. Capobianco, M. Carotenuto, T. Caruso, A. Peluso, The charge-transfer band
1356–1365. of an oxidized Watson–Crick guanosine–cytidine complex, Angew. Chem. Int.
[81] M.-J. Lee, Ch-R. Park, A.-Y. Kim, B.-S. Kwon, K.-H. Bang, Y.-S. Cho, M.-Y. Jeong, Ed. 48 (2009) 9526–9528.
G.-J. Choi, Dynamic calibration for the in-line NIR monitoring of film thickness [96] J.G. Henriques, S. Buziol, E. Stocker, A. Voogd, J.C. Menezes, Monitoring
of pharmaceutical tablets processed in a fluid-bed coater, J. Pharm. Sci. 99 mammalian cell cultivations for monoclonal antibody production using near-
(2010) 325–335. infrared spectroscopy, Adv. Biochem. Eng./Biotechnol. 116 (2009) 73–97.
[82] M.-J. Lee, D.-Y. Seo, H.-E. Lee, I.-C. Wang, W.-S. Kim, M.-Y. Jeong, G.J. Choi, In [97] C. Kurz, M. Leitenberger, R. Carle, A. Schieber, Evaluation of fruit authen-
line NIR quantification of film thickness on pharmaceutical pellets during a ticity and determination of the fruit content of fruit products using FT-NIR
fluid bed coating process, Int. J. Pharm. 403 (2011) 66–72. spectroscopy of cell wall components, Food Chem. 119 (2010) 806–812.
[83] C. Cahyadi, A.D. Karande, L.W. Chan, P.W.S. Heng, Comparative study of non- [98] A. Weber, V. Hoefinfhoff, V. Siekmann, P. Turecek, Detection of physiologi-
descructive methods to quantify thickness of tablet coatings, Int. J. Pharm. cally acceptable polymer molecules using near infrared spectroscopy, United
398 (2010) 39–49. States Patent Application, US 2010/0152115 A1 (2010).
[84] M. Saed, S. Saner, J. Oelichmann, H. Keller, G. Betz, Assessment of diffuse trans- [99] C.-F. Mandenius, K. Graumann, T.W. Schultz, A. Premstaller, I.-M. Olsson, E.
mission mode in near-infrared quantification – Part I: The press effect on Petiot, C. Clemens, M. Welin, Quality-by-design for biotechnology related
low-dose pharmaceutical tablets, J. Pharm. Sci. 98 (2009) 4877–4886. pharmaceuticals, Biotechnol. J. 4 (2009) 600–609.
[85] Y. Yang, A. Gupta, A.S. Carlin, P.J. Faustino, R.C. Lyon, Ch.D. Ellison, B. Rothman, [100] M.C. Sarraguça, A. Paulo, M.M. Alves, A.M.A. Dias, J.A. Lopes, E.C. Fer-
M.A. Khan, Comparative stability of repacked metoprolol tartrate tablet, Int. reira, Quantitative monitoring of an activated sludge reactor using on-line
J. Pharm. 385 (2010) 92–97. UV–visible and near infrared spectroscopy, Anal. Bioanal. Chem. 395 (2009)
[86] J. Cruz, M. Blanco, Content uniformity studies in tablets by NIR-CI, J. Pharm. 1159–1166.
Biomed. Anal. 56 (2011) 408–412. [101] V. Bellon-Maurel, E. Fernandez-Ahumada, B. Palagos, J.-M. Roger, A. McBrat-
[87] E. Chevalier, M. Viana, S. Cazalbou, L. Makein, J. Dubois, D. Chulia, ney, Critical review of chemometric indicators commonly used for assessing
Ibuprofen-loaded calcium phosphate granules: Combination of innovative the quality of the prediction of soil attributes by NIR spectroscopy, Trends
characterization methods to relate mechanical strength to drug location, Acta Anal. Chem. 29 (2010) 1073–1081.
Biomater. 6 (2010) 266–274. [102] Feedback on FDA Validation Guideline., Regulatory highlights for
[88] D. Cozzolino, Near infrared spectroscopy in natural products analysis, Planta March–August 2009, organic process research & development, Am.
Med. 75 (2009) 746–756. Chem. Soc. 13 (2009) 842–847.
[89] D. Cozzolino, A review on the use of near infrared spectroscopy for plant [103] G.L. Bosco, L. James, Waters symposium 2009 on near-infrared spectroscopy,
analysis, Med. Plants – Int. J. Phytomed. Rel. Ind. 2 (2010) 1–8. Trends Anal. Chem. 29 (2010) 197–208.

You might also like