You are on page 1of 28

157

11

Formulation Development Concepts

11.1 Preformulation (NDA) (Bassart 2020). Many techniques and instrumenta-


tions are needed during the discovery and preformulation
Preformulation is often used to describe the physical and phase to establish a baseline characteristics profile (physi-
chemical characteristics of substances and mixtures used cal/mechanical, chemical, and biological) for the new API.
in pharmaceutical products. It is part of the research and Preformulation studies are conducted to examine the com-
development (R&D) unit of the pharmaceutical industry. patibility of the API with various common excipients.
Preformulation is used synonymously with physical phar- Among the physicochemical properties of the API that are
macy. It is considered to be part of a larger science known established during preformulation investigations include
as pharmaceutics. Both preformulation and formulation are melting point/freezing point, porosity, bulk volume, bulk
parts of pharmaceutics, and their application in formula- density, bulkiness, equivalent diameters for powder parti-
tion development in the industry is known as the industrial cles, specific surface, angle of repose, powder flowability, dif-
pharmacy. Characterization of chemicals can be done on ferential scanning calorimetry (DSC), thermogravimetric
the molecular, particulate, and bulk levels (Brittain analysis (TGA), viscosity, viscoelasticity parameters, zeta
1999). Nowadays, during the R&D stage of the pharmaceu- potential and physical stability parameters (creaming/sedi-
tical product’s progress, scientists depend heavily on using mentation and flocculation) for dispersed systems, specific
instrumentation to characterize or identify a chemical or a gravity/density, solubility characteristics in water and
final product. Instruments used in research have become various solvents, dissolution profile, disintegration charac-
extremely complicated and similar to a piano and a musi- teristics of the dosage form, partition coefficient, pH, pKa,
cian’s ability to play it, the same instrument can produce surface tension/interfacial tension, Karl Fischer (KF) titra-
bad, good, or excellent data, depending on how it is being tion for water content, water sorption analysis (dynamic
run (Scarlett 2003). The biopharmaceutical industry alone vapor sorption; DVS), X-ray diffraction analysis (XRD,
has reported an expenditure of $173 billion on R&D in 2017 PXRD, or XRPD), polymorphism characteristics/metastable
(Hilleke and Kars 2019). Because of the importance of the crystals, amorphous forms, and solvate forms, Raman spec-
characterization and the production of drug products, the troscopy, infrared spectroscopy and imaging, Fourier-
pharmaceutical and biotechnology industries have become transform infrared spectroscopy (FTIR), among others. For
the number one target for cybercriminals worldwide example, Stagner et al. (2020) investigated the factors
(Douthwaite 2020). involved in the compressibility, compactibility, and tabletibil-
According to recent estimates, only one out of 10,000 com- ity of flufenamic acid: nicotinamide cocrystal (flufenamic
pounds evaluated becomes an approved pharmaceutical acid is an analgesic, anti-inflammatory, and antipyretic
product for a new active pharmaceutical ingredient (API). agent; nicotinamide is a water-soluble vitamin B-3). The
During this process that lasts on average between 10 and cocrystal formation improves the aqueous solubility of flufe-
14 years, an excess of $1 billion is being spent on the pro- namic acid (Fabian et al. 2011). They aimed to study the
duct’s development (Brown et al. 2019). For New Drug latent effect of storage conditions and material compression
Application (NDA) and Biologics License Application parameters on tablet mechanical stress. Their study used var-
(BLA), the Food and Drug Administration (FDA) has ious techniques that included morphological studies on the
approved an average of 100 NDAs and BLAs in the period cocrystal using scanning electron microscopy, the tablet’s
between 2010 and 2018, 38% of which are new molecular mechanical stress measurement, Shapiro analysis, and
entity (NME) types. The NMEs were either a single active Heckel analysis to describe initial particle arrangement and
(BLA and NDA), a biosimilar (BLA), or a combination type fragmentation during compression. Parameters obtained

Integrated Pharmaceutics: Applied Preformulation, Product Design, and Regulatory Science, Second Edition.
Antoine Al-Achi, Mali Ram Gupta, and William Craig Stagner.
© 2023 John Wiley & Sons, Inc. Published 2023 by John Wiley & Sons, Inc.
Companion website: www.wiley.com/go/Al-Achi/IntegratedPharmaceutics
158 11 Formulation Development Concepts

from the analysis were fitted to multivariate analysis and is a FTIR technique. The following is a synopsis of their
latent variable statistical methodologies such as explora- work (Al-Achi et al. 2019):
tory factor analysis, principal component analysis, and
principal component regression. The statistical methods Inclusion complex can enhance the drug solubility,
were used to describe complex hidden main, interaction, stability, and masking smell. Cyclodextrin (CD) is a
and quadratic effects of the storage conditions and the commonly used host molecule for inclusion, with
compression parameters on the cocrystal tablet mechani- three main types of CD, exist, α, β, and γ. They are
cal strength (Stagner et al. 2020). Interestingly, as recently classified based on their chemical structure. The
as 2019, six out of ten product applications in the United derivative of β-CD, (Hydroxypropyl-β-Cyclodextrin;
States were given accelerated approval via priority review, P-β-CD), is widely used in pharmaceutical formula-
fast track, or breakthrough (Thomas, 2020b). For this tions. HP-β-CD works by a dynamic equilibrium
acceleration to be accomplished, pharmaceutical scien- process for inclusion. Nifedipine is a Class II drug
tists have to produce quality measurements in a shorter (Biopharmaceutical Classification System) that
allotted time, often 18–24 months earlier than typical has poor solubility in water, and high permeability
schedules (Browne and Savla 2019). Moreover, the devel- through the cellular membrane. The Phase-
opment of hybrid products under the European Union solubility profile of nifedipine/HP-β-CD complex
guidelines or their corresponding regulations in the US showed an AL type (according to Higuchi and Con-
505(b)(2) (see Chapter 22 for section 505) require higher nors’ method) indicating a complex ratio of 1 : 1. The
technological innovations to achieve the desired better objective of this study was to investigate the pH
therapeutic effectiveness. It was estimated that one out effect on the inclusion process of nifedipine/HP-
of each two New Drug Applications were subjects to 505 β-CD. The results of this study showed that ioniza-
(b)(2) in 2017 (Anonymous 2018). This is in agreement tion pH conditions improved nifedipine solubility
with the pharmaceutical industry is becoming more and in water to a limited extent however it was not as effi-
more patient-centric in its approach to designing newer cient as the unionization pH conditions since union-
dosage forms (Thomas, 2020b). ization conditions produced higher Complexation
The solubility of chemicals in water is one of the main Efficiency (CE) and the stability constant (K1 : 1)
challenges that hinder the chemical process of becoming value. Therefore, maintaining the drug in a union-
pharmaceutical products. Inclusion complexes formed ized form is perhaps a more efficient way of produ-
between the chemical agent and a carrier system can cing inclusion complex with HP-β-CD. In order to
render the chemical more water-soluble. Cyclodextrins characterize the inclusion complex thus formed
are often used to enhance the water solubility of various between the drug and HP-β-CD, a solvent evapora-
substances by creating an inclusion complex. An example tion method was used to prepare the complex which
of this formulation strategy is the use of a cyclodextrin ini- was compared to a physical mixture, pure drug, and
tially discovered by a group of scientists (Professor Valen- pure HP-β-CD. Significant differences were found to
tino Stella and Dr. Roger Rajewski) at the University of exist between the inclusion complex and all the
Kansas (Sarah Motter 2020). This cyclodextrin is known other groups based on the TGA, DSC, ATR, and
as CaptisolÒ (sulfobutylether-cyclodextrin; Ligand Phar- PXRD analysis.
maceuticals) that combines with various drugs to form The results from the TGA, DSC, ATR, and PXRD are
water-soluble complexes (to date, 15 products have been shown below, respectively (Al-Achi et al. 2019).
approved by various world regulatory agencies using Cap-
TGA-DSC test: After TGA run, the four samples
tisolÒ) (Pipkin et al. 2020). An example of a CaptisolÒ-
were subjected to decomposition temperatures.
containing pharmaceutical product is Veklury® (remdesi-
Based on the temperature data the DSC maximum
vir) by Gilead. The drug is an antiviral agent and suffers
temperature should not be set higher than the
from poor solubility in water. The use of CaptisolÒ in this
decomposition temperature. The inclusion complex
formulation in the ratio of 30 : 1 (CaptisolÒ:API) as a lyo-
had completely turned to amorphous form which
philized powder or 60 : 1 as a solution for injection has
was similar to that of pure HP-β-CD, whereas the
facilitated its complete dissolution in water (Pipkin
DSC curve for pure nifedipine is a typical crystalline
et al. 2020).
form curve which had a sharp endothermic peak at
Al-Achi et al. (2019) examined the enhancement of aque-
170 C. The curve of the physical mixture had a
ous solubility of the antihypertensive drug nifedipine by
smaller endothermic peak which indicated the drug
complexing it with hydroxypropyl-β-cyclodextrin. Physical
was partially included into the HP-β-CD.
characterization of the complex was accomplished by mate-
rial science methodologies (TGA-DSC, attenuated total ATR test: The inclusion complex has a strong peak
reflection analysis (ATR), and PXRD analysis). The ATR in 1678 cm−1, this peak specifically reflects -C=O
11.1 Preformulation 159

group, where nifedipine has two ester groups which showed a typical crystalline form that had many
are lipophilic that can be captured inside of the cav- sharp peaks; for inclusion, the crystal form almost dis-
ity of cyclodextrin, the outside of the cavity is hydro- appeared and the position of the two main broaden
philic that have more -OH groups, it has more peak peaks at the same angle compared with pure cyclo-
intensity than pure drug in 3327 cm−1. Additionally, dextrin suggests that nifedipine was included into
for HP-β-CD, hydroxypropyl group connects the the HP-β-CD. For the physical mixture, it has few
cyclodextrin by an oxygen atom which forms an sharp peaks as compared to pure drug and the general
ether group. Once the drug is included inside, the shape of the graph is more similar to pure HP-β-CD.
inclusion complex has a stronger peak compared
From TGA-DSC, ATR, PXRD studies, all the results
to the pure drug in 1022 cm−1. This was specifically
indicate nifedipine molecule can form an inclusion
demonstrated for the ether (C-O-C) group as seen in
complex with cyclodextrin using solvent evaporation
the pure HP-β-CD spectra.
method in the presence of 40% acetonitrile as the sol-
PXRD test: A sharp contrast was observed between vent. (Al-Achi et al. 2019).
the inclusion complex and pure drug. The pure drug

102
Pure HPBCD
Physical mixture
Pure Nifedipine
Inclusion complex
100

98

96
Weight (%)

94

92

90

88
0 50 100 150 200 250 300 350
Temperature (°C) Universal V4,5A TA

TGA: Source: Al-Achi et al. (2019) / Ocimum Scientific Publishers Pty Ltd / CC BY 4.0.

2 Inclusion complex
Pure HPBCD
Physical mixture
Pure Nifedipine

0
Heat flow (W/g)

–2

–4

–6
0 50 100 150 200 250 300
Exo Up Universal V4.5A TA In
Temperature (°C)

DSC: Source: Al-Achi et al. (2019) / Ocimum Scientific Publishers Pty Ltd / CC BY 4.0.
Drug with inclusion complex
101
100
95

90

85 3325.94 cm–1

1679.21 cm–1
80 3327.38 cm–1 1349.22 cm–1
1431.79 cm–1 829.07 cm–1
%T

1495.61 cm–1
75 1226.91 cm–1 793.63 cm–1
1348.86 cm–1 711.95 cm–1
1678.32 cm–1 744.47 cm–1
70
1119.98 cm–1
65 1225.52 cm–1
1021.88 cm–1

60
1022.80 cm–1
55
54
4000 3500 3000 2500 2000 1500 1000 650
cm–1
Name Description
Complex label 011 by PERC Date Monday, May 18 2015
Pure drug label 014 by PERC Date Monday, May 18 2015

ATR: Source: Al-Achi et al. (2019) / Ocimum Scientific Publishers Pty Ltd / CC BY 4.0.

100 Fingerprint zone of NI COM CD

95

90

85

80 1679.21 cm–1 1349.22 cm–1


1431.79 cm–1 1349.24 cm–1
1495.61 cm–1 829.07 cm–1 744.47 cm–1
75 1527.38 cm–1 1348.88 cm–1 1226.95 cm–1 793.63 cm–1
%T

1678.32cm–1 711.95 cm–1


70

65 1119.98 cm–1
1225.52 cm–1 1021.48 cm–1
60

55
1022.54 cm–1
50
49
1755 1600 1400 1200 1000 800 598
cm–1
Name Description
Pure drug label 014 By PERC Date Monday, May 18 2015
Complex label 011 By PERC Date Monday, May 18 2015
Pure CD label 012 By PERC Date Monday, May 18 2015

ATR: Source: Al-Achi et al. (2019) / Ocimum Scientific Publishers Pty Ltd / CC BY 4.0.

1500

1000

500

0
10 20 30 40 50 10 20 30 40

Inclusion complex Physical mixture

1000

500

0
10 20 30 40 50 10 20 30 40

Pure drug Pure HP-β-CD

PXRD: Source: Al-Achi et al. (2019) / Ocimum Scientific Publishers Pty Ltd / CC BY 4.0.
11.1 Preformulation 161

On the biological front, preformulation testing is also Cell culture research is fast developing with new
important to characterize the biological profile of the instrumentation for studying cells in real time. Systems
API when needed. For example, in evaluating novel anti- such as the ones developed by Sartorius/Essen BioScience
microbial peptides (bacteriocin “Garvicin KS; GarKS” (Essenbio.com/oncology; Ann Arbor, Michigan) (e.g.
was identified from Lactococcus garvieae KS154), Thapa IncuCyte® S3 Live-Cell Analysis System) allow research-
et al. (2020) utilized various techniques and instrumenta- ers in the field of oncology to study cancer cells in their
tions in preformulation testing of this new peptide- living environment noninvasively. Phenomena such as
antibiotic. These techniques included the use of apoptosis, angiogenesis, cell invasion/migration, and anti-
high-performance liquid chromatography instruments, body internalization may be detected by these devices. In
determination of the physical stability of the peptides addition to the cell culture studies, in vivo studies are also
by adsorption and aggregation studies, determination of conducted in experimental animals, including toxicologi-
the chemical stability of the novel drug, determination cal studies where applicable. Thus, it is of importance that
of the antimicrobial activities of the peptides, and deter- the pharmaceutical scientist has a broad comprehension
mination of fibroblast cell viability and proliferation (cell in various process and production techniques including
viability and proliferation studies). In addition, statistical a thorough knowledge in statistical analysis (Chapter 10).
analysis methods were used to detect statistical signifi- Also, pharmaceutical scientists should be familiar with the
cance in the data collected. Among the instruments that various current pharmacopeia requirements in particular
were utilized in the preformulation studies were (Thapa those of the United States, European Union, and Japan,
et al. 2020): in order to meet pharmacopoeia standards (Wiggins and


Albanese 2020). Another consideration is that related to
UV–Vis spectrophotometer (UV-2401 PC, Shimadzu, the mode of pharmaceutical production. Although other
Kyoto, Japan). industries such as aerospace, car making, food, and metal

• HPLC system from Shimadzu Corporation (Kyoto, Japan)


that consisted of an auto-injector SIL-9A, a Prominence
products have been employing continuous manufacturing
processes for decades (Smith 2020), the pharmaceutical
LC-20AD pump, a Chromatopac C-R5A integrator, and industry is slowly moving away from one-batch produc-
a Prominence SDP-10A UV detector. A reversed-phase tion (the traditional batch processing) to continuous pro-
column (Inertsil ODS-SP; 5 μm, 4.6 × 150 mm; GL duction, which is recognized to provide a better product
Sciences Inc, Tokyo, Japan) equipped with a Guard Col- quality, according to the FDA. Applications such as con-
umn (Inertsil ODS-SP; 5 μm, 4.0 × 10 mm; GL Sciences tinuous production offer new challenges to formulation
Inc, Tokyo, Japan). scientists, especially those in biopharmaceutical industry

• For the aggregation studies: A dynamic light scattering


instrument (ZetaSizer, Malvern Instruments, Worcester-
in regard to process and equipment robustness operating
over several days to several weeks at a time (e.g. tubing
shire, U.K.) and mechanical pump degradation) (Brooks 2020).

•• For the adsorption studies: HPLC method.


For the chemical stability studies: HPLC method.
Although continuous processing is financially sound,
the industry still perceives that this way of production

• The antimicrobial activities of individual and combina-


tion peptides were evaluated in methicillin sensitive S.
comes with a “higher regulatory risk” tag (Smith 2020).
The following brief discussion describes results from
Aureus MSSA; ATCC 6538) and methicillin resistant S. preformulation studies that were carried out on human insu-
aureus (MRSA, Xen 31; parental strain ATCC33591). lin (HINS) for the purpose of administering HINS orally or
The turbidity (OD600) of bacterial growth was analyzed transdermally. These routes of administration for HINS
to determine the (minimum inhibitory concentration) remain unattainable for various natural obstacles that govern
MIC value for each peptide. the handling of HINS by the body: in the GI tract, the presence

• Cell culture studies: Adult human dermal fibroblasts


(ATCC®PCS-201–012™) were obtained from ATCC
of proteolytic enzymes; transdermally, HINS has high molec-
ular weight and hydrophilic which render the hormone inca-
(Manassas, VA, USA) and cultured in DMEM supple- pable to undergo spontaneous diffusion through the skin.
mented with 10% FBS, 100 U/mL penicillin, 100 μg/mL Faced with these challenges, Al-Achi and collaborates
streptomycin, and 250 μg/mL fungizone (referred to as reported the following findings on HINS (available from
“complete DMEM media”) in tissue culture flasks. The recombinant DNA technology) (Humulin®R; Eli Lilly; Indi-
cells were incubated at 37 C in a humidified atmosphere anapolis, IN; 100 U/mL) from in vitro, cell culture, in situ, and
of 5% CO2 and sub-cultured twice a week (cells in whole experimental animal studies. It should be noted that
passages 3–10 were used for the experiments). (Thapa the physicochemical characteristics of HINS are well known
et al. 2020) and widely reported in medical literature.
162 11 Formulation Development Concepts

• In vitro study on the diffusion of HINS through


polycarbonate membrane (PM) (Al-Achi and Green-
with HINS. In the presence of the soybean extract, insulin
degradation by these enzymes was significantly reduced,
wood 1995): The PM has been used as a physical support and the protection afforded by soybean extract was more
surface for cellular growth in cell culture studies. The pronounced against trypsin and chymotrypsin than that
membranes were selected with pore sizes of 0.2, 1.0, against pepsin (Al-Achi et al. 2006).
and 8.0 μm (membrane diameter of 13 mm). The diffu-
sion of HINS through the membrane depended on the
• Preparation of HINS with carrier systems: Several
carrier systems were chosen to study their suitability to
pore size, pH, and the presence or absence of collagen interact with HINS. These carriers were erythrocyte
on the membrane’s surface. All experiments were carried ghosts (EG) and sonicated erythrocyte ghosts to produce
at 37 C. HINS diffusion was optimum at alkaline pH and erythrocyte vesicles (EV); some of these membranes were
reached a minimum diffusion point near its isoelectric further coated with liposome-type film (LEG and LEV).
point (pH ≈ 5.3). The presence of collagen on PM’s sur- All carriers were found to associate with HINS, with
face slowed the rate of HINS diffusion and resulted in EG exhibited the highest percent of this association
a lesser total amount of HINS to diffuse over 24 hours. (approximately 50% of the initial amount of HINS was
Larger pores of PM (8.0 μm vs. 0.2 or 1.0 μm) resulted found associated with EG following a 24-hour incubation
in a more significant amount of the hormone diffused period at 37 C) (Al-Achi and Greenwood 1993a). In addi-
(Al-Achi and Greenwood 1995). Moreover, HINS precipi- tion, the interaction of HINS with soybean powder
tated out from its solutions when the pH was between 4 (Glycine max) was investigated. The maximum amount
and 5.8. The diameter of precipitated HINS particles of HINS that can be adsorbed on was experimentally esti-
increased as the pH increased from 4 to 5.8 and reached mated to be 106 U/g of powder. The interaction between
its maximum diameter at pH 5.8 (Al-Achi et al. 1998). HINS and soybean powder was relatively weak, which

• Cell culture studies on HINS diffusion through a


layer of Caco-2 cells (Greenwood and Al-Achi 1997):
allowed easy dissociation of the hormone upon contact
with aqueous media (Al-Achi et al. 2003).
Caco-2 cells are known to undergo a spontaneous differ-
entiation (15–21 post-incubation) in cell culture to enter-
• Preparation of the dosage forms (HINS suspen-
sion): HINS solution was added to soybean extract (see
ocytic cells. This human colon adenocarcinoma cell line above) in a ratio of soybean extract:HINS solution
is commonly used as an experimental model to study (100 U/mL) 1 : 9, and the pH of the mixture was adjusted
drug diffusion through the intestinal epithelium. The to 4.0. At this pH, HINS precipitated from solution forms
cells were grown on the surface of polycarbonate-colla- a suspension with a dispersed phase average diameter
gen-coated membranes (Corning® Transwell® polycar- (dvs) of 29.93 μm. The shelf life of the HINS suspension
bonate membrane cell culture inserts) at a density of was estimated to be approximately 18 days at 4 C
60,000 cells/cm2. The permeability coefficient of HINS (Al-Achi and Parekh 2004).
through a differentiated layer of Caco-2 cells was (6.5 ±
1.2) × 10−7 cm/s. This permeability coefficient was similar
• Diffusion of HINS through excised skin: These experi-
ments were done using excised hairless mouse skin. The
in value to that reported elsewhere from an everted rat skin was mounted on Franz cell apparatus, and the diffu-
small intestine model (Greenwood and Al-Achi 1997). sion of HINS through the skin with or without absorption

• In vitro studies on the stability of HINS: Soybean


powder (mean volume-surface diameter dvs = 45.5 μm;
enhancers [limonene solution in alcohol (5%), iodine tinc-
ture, and ethyl acetate in alcohol (1 : 1)] was documented.
average bulk volume 44.5 mL; average bulk density Limonene solution in alcohol facilitated the diffusion of
0.625 g/mL; average angle of repose 38.33 ; and average HINS, the most compared with the other two enhancers
Carr index 26.72%) was prepared by grinding the beans (the permeability coefficient of HINS through the skin
(Glycine max) in a coffee grinder (Mr. Coffee, Sunbean was 9.47, 4.42, and 2.78 × 10−6 cm/s for limonene, iodine,
Products, Hatiesburg, MS), and an aqueous extract and ethyl acetate, respectively). The pretreatment with
(pH = 6.5–7.0; density = 1.0178 g/mL; viscosity = 1.39 limonene alcoholic solution produced a HINS flux
centipoises) was prepared from the resulting soybean through the skin of 1.23 U/cm2/h, which imitated the pan-
powder (Al-Achi et al. 2003). The aqueous extract was creatic normal secretion rate. There was no HINS diffu-
prepared by mixing 5 g of the powder with 25 mL water sion through the skin when the hormone was applied
and the mixture was incubated for one hour at 37 C. The without an absorption enhancer (Al-Achi et al. 2011).
mixture was then centrifuged and filtered to produce a
clear extract (Al-Achi et al. 2006). Soybean contains sub-
• In situ experiments (Al-Achi and Greenwood 1993b):
HINS was administered intraduodenally to anesthetized,
stances in its protein fraction with anti-proteolytic activ- streptozocin-induced diabetic rats. EG, EV, LEG, and
ities, inhibiting trypsin and chymotrypsin from reacting LEV HINS-carrier systems and the free hormone were
11.1 Preformulation 163

administered locally, and the blood glucose of rats was to the thepharmaletter (https://www.thepharmaletter.
monitored over five hours. Free HINS did not produce com/article/growth-forecast-for-drug-delivery-systems-
any hypoglycemic activities, while HINS-carrier systems market (accessed 14 September 2020)), due to a forecasted
showed varying degrees of hypoglycemia; LEV was the rise in future infections that require specialized instrumen-
most effective carrier for delivering HINS to the circula- tations (e.g. ventilators), CAGR for drug delivery systems
tion via the intraduodenal route as indicated by a pro- market is expected to be at 7% between 2020 and 2025.
nounced hypoglycemic state (Al-Achi and Greenwood Another driving force behind the rise in predicted growth
1993b). The LEV-HINS carriers were further investigated in drug delivery technologies is the extension of a product’s
and found to be equally effective (i.e. causing hypoglyce- life while minimizing cost (Hamilton and Lutz 2005).
mic effect) at doses of 200, 400, or 600 U/kg body weight. Development of a new delivery system for a given drug
Interestingly, the administration of free HINS via this begins in research and development (R&D) departments
route in high enough doses (600 U/kg body weight) also in the pharmaceutical industry, utilizing the principles of
produced a hypoglycemic response (Al-Achi and Green- preformulation. However, there is no guarantee whatso-
wood 1994). ever that a specific formulation that was generated in

• Whole experimental animal studies (Al-Achi and


Greenwood 1993c): HINS free or associated with the car-
R&D will be applicable in the production area. In addition,
the pharmaceutical industry utilizes what is known as proc-
riers mentioned above were administered either orally ess analytical technologies (PATs). The main purpose of
(by gavage) or buccally in streptozocin-induced diabetic PAT is for control and assessment during manufacturing.
rats. The latter form of administration resulted in a low- For instance, the compression force signal during tableting
ering in blood glucose level following free HINS or EG- can be used to accept or reject a tablet during processing
HINS. These findings led to the conclusion that HINS (Manley et al. 2019). Moreover, near-infrared spectroscopy
could pass through the buccal mucosa, reaching the cir- is being employed as PAT for many steps in tablet manufac-
culation to affect a state of hypoglycemia (Al-Achi and turer such as blending uniformity, as well as for moisture
Greenwood 1993c). Expect for LEG-HINS, which was content detection in the finished tablets (Rajan et al.
ineffective in lowering blood glucose level, the oral 2010). It is important for the R&D department and the pro-
administration of the other carriers-HINS mentioned duction operation facility to coordinate their use of PAT
above or free HINS (333 U/kg of rat body weight) resulted and to allow communication concerning PAT to flow
in a hypoglycemic response (Al-Achi and Greenwood smoothly back and forth (Arrivo 2003). The obvious advan-
1998). This response has demonstrated once again that tage of PAT is that it provides online, in-place analysis for
a large enough oral HINS dose could bring down blood an active pharmaceutical ingredient (API) during the pro-
glucose level, even when HINS was in its free form in cessing steps, eliminating the need for repeated sampling
solution. The oral administration (by gavage) of soybean during the manufacturing operation. Thus, it reduces the
extract-HINS suspension (1 mL soybean extract was com- exposure of personnel to APIs, a matter of importance
bined with 1 mL of Humulin®R; 167 U/kg of rat body when handling highly potent drugs. PAT also reduces both
weight) in diabetic rats resulted in a significant reduction the overall cost and the time of operation (Arrivo 2003). In
in blood glucose level that was sustained for four hours September 2004, the US FDA issued a Guidance for Indus-
post-administration. The lowering in blood glucose was try document concerning PAT. The use of PAT allows the
observed as early as 30 minutes following the oral dose. industry to produce a pharmaceutical product of high qual-
In contrast, control groups (saline or soybean extract) did ity in a consistent and predictable manner (Dziki 2008).
not show any hypoglycemic effects (Al-Achi et al. 2012). The purpose served by R&D is often questioned, since a
The administration of a 200 U/kg of rat body weight considerable amount of information learned there may not
HINS oral dose to diabetic rats did produce any reduction be applicable to production. In this chapter, we shed some
in blood glucose either (Al-Achi and Greenwood 1994). light on issues related to transferring a formulation from
R&D to production (i.e. the scale-up process), rate-
It has been stated that only 1 out of 11 drugs entering controlled drug delivery systems, novel drug delivery
phase 1 clinical investigations reach the market, and only technologies to enhance oral bioavailability, possible
one-third of the drugs that make it to the market cover their drug–excipient or excipient–excipient interactions, con-
development cost (Dubin 2006). It was also predicted that tamination issues, and other issues. To make the point in
the market for drug delivery systems would grow at an another way, according to Arthur Calder-Marshall, “out
annual rate of 10%, reaching $132 billion in 2012 (Dubin of sight, out of mind” translated into Russian by a com-
2009). A compound annual growth rate (CAGR) of 4.9% puter, then back into English, became “invisible maniac”
for advanced drug delivery technologies was expected (Tingstad 1988). Thus, clear and open communications
between 2015 and 2020 (Saharan 2019). And, according between a R&D department and production personnel
164 11 Formulation Development Concepts

cannot be overemphasized for the ultimate success of the inlet air (Gore et al. 1985; Mehta 1988). Granulations made
final product. by high-shear mixtures present another type of challenge
Eight principles determine whether or not a process is suit- for scale-up. There appear to be four areas of concern
able for manufacturing a drug product. The process must be regarding this scaling-up operation: the first three being
robust, simple, safe, standardized, scalable, minimize the variations in the API and the excipients, in-process con-
cost and time of production, and transferable/scalable trols, and failure to recognize the fact that scaling-up for-
(Brower et al. 2020). mulations from R&D to production is not a linear
process. However, the most significant factor affecting
the process of transition to large-scale production was
found to be the variation encountered with the API
11.2 Scale-up Considerations (Chowhan 1988).
Film coating is another area of concern during scale-up.
Issues related to scaling-up dosage forms from the R&D For film coating, two major types of equipment are availa-
stage to the pilot stage and thereafter to production are ble: fluid-bed type and pan coaters. Each of these coaters
numerous and often lead to failure in producing the exists in many types, and those used in the R&D stage
planned dosage form unless addressed and resolved. Most are obviously smaller than those employed in production.
problems encountered during scale-up are related to solid Smaller instruments handle materials in a much gentler
dosage forms such as tablets and suppositories (Russo way. Large production large equipment could damage a
1984; Chowhan 1988), as well as lyophilization techniques product at any step before and after coating (Pondell
(Tchessalov et al. 2007). Variability in the chemicals with 1985). Therefore, care must be exercised to protect a prod-
respect to their physical characteristics and equipment var- uct during handling in the production areas, despite evi-
iability are perhaps the main areas of concern during scale- dence of “stability” throughout the R&D process. Other
up (Russo 1984; Pondell 1985). For example, during tablet factors that may contribute to the quality of the finished
formulation, variations exist during milling and mixing, coating film include the solid content of the coating disper-
and in the types of presses used (Russo 1984). Moreover, sions, the number of spray guns used, the pan speed (in the
a fluid-bed granulation process is often used in industry case of pan coating devices), and the coloring agent if used
in tablet manufacturing. The basic fluid-bed granulation in the coating dispersion. All these factors play a significant
unit consists of an air-handling unit, an exhaust air turbine, role during processing of the final product formulation
an exhaust air filter, a processing zone (i.e. dryers, granula- (Porter and Saraceni 1988).
tors, spray nozzles), and product discharge components Several drying technologies are utilized in industry for
(Olsen 1989). This technique allows the production of gran- the purpose of improving product’s stability. These technol-
ules with ideal features of spherical shape, high porosity, ogies include lyophilization (freeze-drying via sublimation
and narrow size range. These features provide excellent of water), spray drying, and reduced-pressure drying
flow characteristics and compressibility for the resulting (Kawasaki et al. 2019). While freeze-drying techniques
granulation. Production batches are naturally higher in are useful for a variety of products, including biologicals,
weight and require more time for processing. Fluid beds spray-drying techniques are limited to low-molecular-
employed in the production stage produce higher shearing weight compounds due to the heat and sheer-stresses used
forces to form the desired agglomeration. They utilize during spay drying cycles (Lowe et al. 2018). Lyophilization
warmer fluidizing air than that used with R&D units, is a commonly employed process in industry for the prep-
and the rate for delivering the binder dispersion is many- aration of sterile powdered dosage units. This technique
fold higher than that of a R&D application. These two fac- requires several steps that may create challenges during
tors present in the production unit produce granulation the scale-up process. Lyophilization process involves three
containing a lower moisture content just prior to the drying main stages; these are freezing stage and the formation of
stage. Production tablets often have higher disintegration ice crystals, primary drying stage (sublimation of ice crys-
times, albeit not significant enough to affect the dissolution tals; it is the longest of the three stages), and secondary dry-
profile of tablets. To achieve a smooth transition from R&D ing stage (desorption of unfrozen water) (Kawasaki et al.
to production utilizing fluid-bed technology, factors such as 2019). Several instrumentations may be used to monitor
binder delivery rate and moisture content must be consid- the temperature and pressure during the freeze-drying
ered (Gore et al. 1985). Overall, and for the fluid-bed gran- cycles. These monitoring devices include capacitance man-
ulation scaling-up procedure, close attention should be ometers, Pirani gauges, resistance temperature detectors,
directed to factors such as batch size, type of equipment and thermocouples (Sacha 2020). In addition to these chal-
used, delivery rate of binders, pressure of the atomization lenges faced during cycle development, formulators have
air, volume of the fluidization air, and temperature of the also to consider other areas of the freeze-drying process
11.2 Scale-up Considerations 165

such as cake characterization and process optimization systematic approach to development that begins with pre-
(Zhang et al. 2020). From the point of view of the scal- defined objectives and emphasizes product and process
ing-up technique, temperatures during the freezing, pri- understanding and process control, based on sound science
mary, and secondary drying steps must be similar for the and quality risk management” be implemented (Kawasaki
laboratory and manufacturing processes. This guarantees et al. 2019). The guiding principles of QbD along with their
not only the same product characteristics but also the same attributes related to lyophilization process are summarized
stability profile (Tchessalov et al. 2007). Thus, two para- in Table 11.1 (Kawasaki et al. 2019).
meters for a successful scale-up of the lyophilized product However, what QbD principles apply to small-molecule
have emerged for their importance in this respect; these are production may not necessarily be fully applicable to bio-
the water vapor transfer resistance of the dried layer that is pharmaceuticals. These latter products have significantly
formed during the freeze-drying cycle and the vial heat more quality attributes than those encountered with small
transfer coefficient. The latter is the algebraic sum of three molecules (Menezes et al. 2019). An example of these appli-
other coefficients, namely the contact heat transfer, the gas cations to biopharmaceuticals is applying QbD principles to
heat transfer, and the radiant heat transfer coefficients develop a high-dose, self-injectable delivery system of bio-
(Kawasaki et al. 2019). The vial heat transfer coefficient logics where the need to inject a viscous liquid with a high
value is directional proportional to the mass loss over time volume (2 mL or more) via an auto-injector exists. For such
and the latent heat of ice and inversely proportional to the a case, a patient-centric approach is needed that provides
cross-sectional area of vial (calculated from its outer diam- robustness and acceptability by the user (Le Gal 2019).
eter) and the difference between the shelf surface temper- Lyophilization can also be applied to liposomal type for-
ature and the product temperature. The water vapor mulations. This is done to improve the shelf life of the lipo-
transfer resistance is inversely proportional to the mass loss somal preparation. The challenges that face the formulator
over time and directly proportional to the cross-sectional in this process are related to the freezing and the dehydra-
dried layers and the difference between the equilibrium tion of the liposomes that lead to destabilization of the
vapor pressure of ice and the vacuum pressure (Kawasaki vesicular structure with the concomitant drug leakage
et al. 2019). (Franzé et al. 2018). Experimental data have shown that
At the beginning of the twenty-first century, the transfer substances such as fructans (inulins; linear fructose poly-
of a lyophilized product from R&D to production necessi- mers; isolated from plants such as chicory roots and dahlia
tated that the production team relied on trial-and-error sort tubers), when combined with glucose are capable of stabi-
of approach to produce the lyophilized product in industrial lizing the phospholipid structure of liposomes during the
quantities. This was dramatically changed, toward the end lyophilization (Hincha et al. 2000). Others (Kannan et al.
of the first decade of this century, with the establishment of 2015; Guimarães et al. 2019) have found that the inclusion
the principles of quality-by-design (QbD) with its critical of sucrose as a lyoprotectant during the lyophilization proc-
elements, namely the Design Space and Process Analytical ess of liposomes has greatly improved the physical stability
Technology (PAT). These principles indicated that “a of the formulation. It is important to note here that glucose

Table 11.1 QbD: guiding principles.

Lyophilization parameters
Guiding principles Description or attributes

Critical Quality Physical, chemical, biological, or microbiological properties or


•• Glass transition temperature

••
Attributes (CQAs) characteristic that should be within an appropriate limit, range, or Eutectic temperature
distribution to ensure the desired product quality Cake collapse temperature


Product temperature
Water vapor transfer resistance of
the dried layer

Critical Process Process parameters whose variability has an impact on a CQA and •• Freezing temperature

••
Freezing rate
Parameters (CPPs) therefore should be monitored or controlled to insure the process produces
Annealing temperature/time
the desired quality
Primary and secondary drying
temperature, pressure, and time

Critical Material Attributes of input materials whose variability has an impact on a CQA and •• Related substances

••
Appearance
Attributes (CMAs) therefore should be monitored or controlled to insure the process produces
Water content
the desired quality
Reconstitution time
Source: Adapter from (Kawasaki et al. 2019).
166 11 Formulation Development Concepts

and sucrose exhibit a collapse temperature (Tc) and glass B virus. Examples of the vaccines that are approved for
transition temperature of maximum freeze concentrate treating cancer are sipuleucel-T (Provenge®; Dendreon
(T g) of (−40 C; −43 C) and (−32 C; −32 C), respectively Pharmaceuticals LLC, for the advance stage of prostate
(Franzé et al. 2018). If the prevailing temperature is below cancer – metastatic castration-resistant prostate cancer)
T g, the material resembles solid glass; and when the ambi- and talimogene laherparepvec (Imlygic®; Amgen; for mel-
ent temperature is below Tc, the material can support and anoma of the skin and lymph nodes) (Baluja 2018). It has
maintain its “rigid” structure. Other commonly used lyo- been reported that by mid-2017, there was an estimated
protectants during the lyophilization process include total of 240 new vaccines and immunotherapies against
(Franzé et al. 2018) arginine, dextrans (various molecular cancer being investigated in clinical trials (O’Connor
weights), gelatin, histidine, HP-β-cyclodextrin, kaemp- 2019). Vaccines that are used to fight infectious diseases
ferol-3-O-glucoside, lactose, lysine, maltoexaose, malto- are defined as antigenic substances that are made from
heptaose, maltose, maltotetraose, maltotriose, mannitol, the causative agent of a disease or a synthetic substitute
quercetin-3-O-glucoside, quercetin-3-O-rhamnoside, and or carry a genetic material (e.g. mRNA) of the infectious
trehalose. It should be noted that the lyoprotectants vary agent to promote the making of antigens by the host cells
in their effectiveness with the type of liposomes being used that trigger the immune system to build antibodies against
(Franzé et al. 2018; Guimarães et al. 2019). them (e.g. vaccines made by Pfizer-BioNTech and Moderna
As stated above, the main advantage for lyophilization is against COVID-19 viral infection, caused by SARS-CoV-2).
to render the product more stable than if it were in a liquid Vaccines are used to provide immunity against one or mul-
dosage form. Thus, lyophilization techniques require low tiple diseases (Calle 2020). This immunity is often granted
temperatures during processing, which is highly suited to early in life during childhood. Pediatric vaccines are many
heat-sensitive APIs. In addition, from the cost point of view, and include conjugate, live-attenuated, toxoid, and inacti-
it is cheaper to transport lyophilized units than liquid-filled vated vaccines (Wei Ling 2020). Some of the vaccine formu-
containers due to weight differential between the two dos- lations may also contain substances that are known
age products (Thomas 2020a). It should be noted, however, collectively as “adjuvants.” According to the US FDA, an
that not all formulations can be subject to lyophilization. adjuvant is “a substance added to some vaccines to enhance
For instance, those containing high salt concentration have the immune response of vaccinated individuals.” The FDA
a reduced freezing point (based on the colligative properties lists several of those adjuvants that are found in some of the
of solutions) and are difficult to freeze-dry. Moreover, for- vaccine preparations. The most important of which are alu-
mulations with volatile components that have high vapor minum salts, monophosphoryl lipid A, an oil-in-water
pressure (e.g. acetate buffers) and can be lost during the emulsion composed of squalene, a combination of mono-
vacuum cycle. Techniques, such as lyophilization, also phosphoryl lipid A and QS-21 (a natural compound found
require highly trained pharmaceutical scientists specializ- in the central Chilean evergreen soapbark tree Quillaja
ing in this area of formulation applications (Thomas saponaria Molina), AS04, which is a combination of alumi-
2020a). Such expertise is available at companies that spe- num hydroxide and monophosphoryl lipid A, and CpG
cialize in this type of technology. Exelead (Indianapolis, 1018 (cytosine phosphoguanine – a synthetic form of
IN) is such a company that offers supports in developing DNA, which resembles the genetic composition of bacteria
liposomal and PEGylation formulations for parenteral ster- and viruses). Vaccines that do not need adjuvants for their
ile drug products (in areas such as enzyme replacement activation include the messenger ribonucleic acid (mRNA)
treatment, infectious diseases, and oncology). Incidentally, types. The mRNA vaccines rely on introducing a synthetic
PEGylation is the process of attaching a polyethylene oxide viral mRNA to the recipient’s cells, allowing the cell to man-
polymer to a pharmacologically active drug molecule. The ufacture viral proteins. These, in turn, are recognized by the
purposes of which are to extend the biological half-life of organism as antigens, and the immune system raises antibo-
the drug and, more importantly, to diminish the host’s dies against them. Multiple dosing is commonly needed with
immune responses. Overall, PEGylated drug molecules the mRNA vaccinations to achieve their desired protective
have better therapeutic actions (Okutgen et al. 2020). effect against viral infections. Recent examples of mRNA
vaccines are the ones developed by Pfizer-BioNTech (New
York, NY) and Moderna (Cambridge, MA) to combat novel
11.2.1 Vaccines
coronavirus infection (COVID-19). The active ingredient in
The primary purpose of administering vaccines is to stim- the Pfizer COVID-19 and Moderna vaccines is nucleoside-
ulate the immune system to identify and protect the indi- modified mRNA encoding the viral spike (S) glycoprotein
vidual from impending infections. Some vaccines are of SARS-CoV-2. They differ in their inactive ingredients;
made to fight cancer development or to help in its treat- the Pfizer-BioNTech vaccine contains 2[(polyethylene glycol
ment. Examples of the formers are the vaccine used against (PEG))-2000]-N,N-ditetradecylacetamide (protects/shields
human papillomavirus and another against hepatitis mRNA from cell’s microenvironment and lipid lubricant
11.2 Scale-up Considerations 167

to facilitate cell entry), 1,2-distearoyl-sn-glycero-3-phospho- given orally, they also tend to interact with the lymphatic
choline, (4-hydroxybutyl)azanediyl)bis(hexane-6,1-diyl)bis system elements populating the digestive tract pathway,
(2-hexyldecanoate) (protects/shields mRNA from cell’s which leads to activation of the immune system quicker
microenvironment and lipid lubricant to facilitate cell entry), than that observed from a parenteral dose administration
sodium chloride (stabilizer), potassium chloride (stabilizer), (Vela Ramirez et al. 2017). Vaccines may be dispensed as
and dibasic sodium phosphate dehydrate (pH adjustment). a single-unit container or a multi-dose container. For the
Those in the Moderna vaccine are PEG2000-DMG: 1,2- multi-dose containers, a preservative is needed to prevent
dimyristoyl-rac-glycerol (protects/shields mRNA from cell’s bacterial growth within the product over its shelf life.
microenvironment and lipid lubricant to facilitate cell entry), A list of licensed vaccines in the United States is found
methoxypolyethylene glycol (protects/shields mRNA from on the FDA website (https://www.fda.gov/vaccines-
cell’s microenvironment and lipid lubricant to facilitate cell blood-biologics/vaccines/vaccines-licensed-use-united-
entry), 1,2-distearoyl-sn-glycero-3-phosphocholine (pro- states (accessed 9 September 2020)). Vaccines for viral
tects/shields mRNA from cell’s microenvironment and lipid infections can be prepared with live attenuated virus, inac-
lubricant to facilitate cell entry), SM-102: heptadecan-9-yl tivated virus, or by recombinant methods. Those made to
8-((2-hydroxyethyl) (6-oxo-6-(undecyloxy) hexyl) amino) protect against a bacterial infection are prepared from
octanoate (protects/shields mRNA from cell’s microenviron- whole-cell inactivated bacteria, bacterial sub-units, toxoid,
ment and lipid lubricant to facilitate cell entry), trometha- or conjugate forms (Plotkin et al. 2017).
mine (pH adjustment), tromethamine hydrochloride (pH Scale-up of vaccines (Markarian 2020) to produce mil-
adjustment), acetic acid (pH adjustment), and sodium lions or even billions of units can be a magnificent chal-
acetate (pH adjustment). They both contain cholesterol lenge. The cost for establishing a new vaccine facility is
(protects/shields mRNA from cell’s microenvironment and estimated to be in the hundreds of millions of US dollars
lipid lubricant to facilitate cell entry) and sucrose (cryopro- for mammalian-cell-based facilities (Robinson 2009).
tector). The Johnson & Johnson/Janssen vaccine against Depending on the medical urgency, such as during pan-
COVID-19 is based on viral vector technology and has as demics, the units to be produced occasionally have to be
its active ingredient the recombinant, replication- manufactured at different locations and within a short
incompetent Ad26 vector, encoding a stabilized variant of period of time. Thus, in addition to controlling for the
the SARS-CoV-2 Spike (S) protein. The vaccine formulation raw materials quality (e.g. containers) and manufacturing
also contains inactive ingredients: Polysorbate-80 (Tween- procedures (e.g. maintaining sterility during processing),
80; surfactant/stabilizer), 2-hydroxypropyl-β-cyclodextrin issues related to training personnel at the different facilities
(solubilizer/stabilizer), citric acid monohydrate (pH adjust- to get them to the same level of competency is an added
ment/stabilizer), trisodium citrate dehydrate (pH adjust- consideration for the production team to plan for
ment/stabilizer), sodium chloride (stabilizer), and ethanol (Markarian 2020). Also, for manufacturers with other pro-
(solvent). All three vaccines are free from eggs, preservatives, ducts being manufactured within the same facility, this
latex, and metals (iron, nickel, cobalt, lithium, rare earth raises the question of cross-contamination. In situations
alloys, or any manufactured products) (https://www.cdc. like these, the manufacturer has to have “cleaning-in-
gov/vaccines/covid-19/clinical-considerations/covid-19- place” procedures for all surfaces, including those of the
vaccines-us.html#Appendix-C (accessed 16 August 2021)). equipment that is used for the end units, as well as for
Another relatively new development in this field is that the intermediate products (Challener 2020). Of course,
the FDA granted Seqirus Inc. (Maidenhead, UK) authoriza- the issue of cross-contamination can be greatly minimized
tion in February 2020 to market Audenz (influenza A/ or totally eliminated by the use of single-use platforms such
H5N1 monovalent vaccine for persons six months of age as the automated one available from Sartorius (High Perfor-
and older). This approval would be the first time that the mance Single Use Platform; BIOSTAT STR Generation 3)
FDA has permitted a cell-based, adjuvanted (adjuvant for bioreactors (Sartorius, Goettingen, Germany). This sys-
was MF59®) vaccine in the United States. The company tem provides real-time data collection and it is data-driven;
is producing the vaccine in its facility in the US (Holly the system is completely controlled by BIOBRAIN, an auto-
Springs, NC). mation platform. Pall Corporation (a global company) has
Some of the available vaccines are given via parenteral introduced the iCELLis bioreactor system that offers an
administration, and some are administered orally. The oral automation platform with a single-use capability, which
route of administration offers the advantage of being safer greatly facilitates the scale-up of vaccine manufacturing
and easier to deliver than the injectable forms, leading to (Castillo 2013). And the demand for these types of reactors
improving compliance. From the manufacturing point of is on the increase (13.5% per year) (Mirasol 2020). Some of
view, it is relatively easier, in general, to produce an oral these new bioreactors have double the upper range capacity
dosage form than a parenteral one. When vaccines are of 2000 L, reaching upward to the 6000 L volume capacity
168 11 Formulation Development Concepts

(experts argue over their usefulness beyond the current VACCINE (TICE® strain) (for the prevention of tuberculo-
2000 L bound) (Mirasol 2020). It should be noted that, in sis; Organon Teknika Corporation LLC, Durham, NC). A
general, single-use platforms are more suited for persona- list of licensed vaccines in the United States is found on
lized cell/gene therapy, while continuous processing appli- the FDA website (https://www.fda.gov/vaccines-blood-
cations are more utilized in large production volume biologics/vaccines/vaccines-licensed-use-united-states
methodologies, such as vaccine or virus manufacture (accessed 14 September 2020)).
(Anonymous 2020a). Although the development of a new Manufacturing platforms for vaccine production vary greatly
vaccine is highly time consuming, Novavax, Inc. among pharmaceutical companies. This variability in the
(Rockville, MD) has developed a rapid system for preparing production is due to the different forms of vaccines that can
influenza vaccine that is an insect-cell based (a new vaccine be produced (active vs. inactive form; parenteral vs. oral admin-
for influenza can be made within 10–12 weeks). This insect- istration). Table 11.2 summarizes some of the substances found
based system allows the use of disposable reactors due to its in vaccine products licensed in the United States (https://www.
high yield production (Robinson 2009). In 2020, Novavax, fda.gov/vaccines-blood-biologics/safety-availability-biologics/
Inc. platform was utilized to produce a vaccine against common-ingredients-us-licensed-vaccines (accessed 10
the novel coronavirus (COVID-19) pandemic. At the writ- September 2020)).
ing of this section, a dozen of other companies are engaged
in developing vaccines against this virus that was discov-
ered in China during the fall season of year 2019. The virus
spread worldwide and has resulted in hundreds of thou- 11.3 Combination Products
sands of people dying from the viral infection globally.
COVID-19 is particularly dangerous in patients with Compliance with taking medications is a huge issue in clin-
comorbid conditions (e.g. diabetes, obesity, high blood ical practice. It is estimated that the cost of noncompliance
pressure) and seniors. For the latter group, their immune can be as high as $100 billion annually, accounting for addi-
system is weakened with aging, and a supportive-type vac- tional health care expenditures and lack of productivity
cine in addition to the leading vaccine may be of impor- (Dubin 2005a). Combination products offer one solution
tance to provide an added prolonged immune support to improving patient compliance. The notion of combina-
through T-cell-mediated immunity (Wolf 2020). One of tion products is not a new concept in the pharmaceutical
the major challenges that face vaccine development against industry. A quick perusal of clinical reference texts reveals
viruses is the ability of the virus to mutate. For instance, their presence since the industry began. However, the old
Influenza A has 18 different subtypes and thousands of types of combination products were limited to certain drugs
strains for each subtype. By mutating, the virus can go that did not show compatibility issues, and their quantities
undetected by the immune system (Panjwani 2019). were small enough to accommodate them within a single
Vaccine production is licensed for the actual biological dosage unit. The new combination products are much more
entity and for the manufacturing process for producing versatile in their ability to include products in larger quan-
the final product. Changes in the manufacturing process tities and regardless of compatibility issues. Multicompart-
can trigger licensing-related issues with regulatory agencies ment capsules (e.g. InnerCap Technologies, Inc; Tampa,
(Plotkin et al. 2017). In manufacturing vaccines, several FL) are made to house several drugs to treat the same or
considerations have to be identified for producing the final different conditions, regardless of their compatibilities.
product. These considerations include (1) biological aspects The new capsules are made of a suitable material, such
such as the nature of the microorganism, (2) growth as hydroxypropyl methylcellulose, gelatin, starch, or a com-
medium aspects (the specifications of the microbial cul- bination. Coating materials can be applied to the outer
ture), (3) the phases needed for the purification processes, walls for specially targeted delivery. [Unlike hard-shell cap-
(4) personnel-related knowledge and experience of the sules, soft gelatin capsule shells are to date only available as
manufacturing process, and (5) the methods used to ana- gelatin-based units (Thomas 2020b).] Another example of
lyze the antigens and other biological processes resulting multicompartment capsules is BilDil (NitroMed Inc., Lex-
from vaccine production (Plotkin et al. 2017). Stabilization ington, MA), containing the combination vasodilator drugs
of the vaccine preparation can be done by preparing the isosorbide dinitrate and hydralazine hydrochloride
solution under optimal pH for stability using a buffer sys- approved by the FDA for the treatment of heart failure in
tem or preparing a lyophilized dosage form that is less African-American patients (Dubin 2005b).
prone to degradation (Plotkin et al. 2017). Examples of vac- In general, combination products can deliver simultane-
cines prepared by lyophilization are VAXCHORA™ ously, within the same unit, a drug, a biological, and a med-
(Cholera Vaccine, Live, Oral), Suspension for Oral Admin- ical device. Most of these products (90%) are currently being
istration (PaxVax, Inc., Redwood City, CA), and BCG developed for cardiovascular applications and focus on
11.3 Combination Products 169

Table 11.2 Substances found in vaccines.

Substance name Function

AS04 (a combination of aluminum hydroxide and monophosphoryl lipid A) Adjuvant


AS03 (D,L-alpha-tocopherol (vitamin E), squalene, and polysorbate 80) Adjuvant
MF59 (an oil-in-water emulsion of squalene oil) Adjuvant
CpG 1018 (synthetic DNA sequencing) Adjuvant
AS01B (a combination of monophosphoryl lipid A and QS-21, a substance Adjuvant
extracted from the Chilean tree Quillaja Saponaria)
Aluminum salts (e.g. aluminum hydroxide) Adjuvants
Neomycin Antibiotic
Polymyxin B Antibiotic
Streptomycin Antibiotic
Gentamicin Antibiotic
Fetal calf/bovine serum Cell nutrients during manufacture
Formaldehyde (≤0.005 mg–0.1 mg/dose) Denaturing agent
Thimerosal (0.001–0.01%) Preservative
Phenol (0.25%) Preservative
2-Phenoxyethanol (100 μg/mL) Preservative
Benzethonium chloride (25 μg/mL) Preservative
Lactose Stabilizer
Sucrose Stabilizer
Glycine Stabilizer
Monosodium salt of glutamic acid Stabilizer
Human serum albumin Stabilizer
Gelatin Stabilizer

developing stent drug delivery (Prajapati et al. 2008). Other (North Brunswick, NJ) has developed nanotechnology plat-
applications involve areas related to surgical dressing and forms suitable for oral and parenteral administration.
orthopedics. Also, an emerging area of combination prod- Ascendia’s EmulSol allows the formation of oil-in-water
uct technology includes nanotechnology (Ford 2008). This nanoemulsions (droplet sizes: 50–500 nm), with drug solu-
area has evolved into a new specialty known as nanomedi- bilized within the droplets of the oily phase. The nanoemul-
cine. Applications in this area include improved sion is formed through homogenization. This method was
antibacterial agent activity, acting as carriers, improvement found helpful in enhancing the bioavailability of drugs and
in implantable materials, diagnostic applications, and sen- allowing medicines that are not candidates for parenteral
sory units in various devices (Prajapati et al. 2008). For administration to be given via this route (e.g. clopidogrel;
example, new technologies are being developed to over- anti-thrombotic) (Huang 2020a). Unlike microemulsions
come issues related to post-stent placement (e.g. re- that are considered to be thermodynamically stable systems
blocking of arteries, thrombosis, blood clots). Among these due to the presence of stabilizers (surfactants and co-sol-
technologies, Micelle technologies’ polymer nanoparticle vents), nanoemulsions are subject to Ostwald ripening
coating technique allows multiple drug delivery from a effect, which tends to destabilize their formulation (see
single stent, as well as controlled delivery of drugs in a Chapter 15) (Huang 2020a). The other platforms of
sequential manner (Andrews and McClain 2008). Ascendia are AmorSol, NanoSol, and controlled-release
A French-government-funded technological research applications. According to Ascendia, AmorSol is for the
organization, CEA-Leti, is currently investigating the use production of amorphous solid dispersions; NanoSol is
of a nanoparticulate system (LipidotsTM; bio-assimilable for producing nano-sized drug particles. Applications for
lipids) for developing a vaccine for human immunodefi- controlled-release techniques are for poorly soluble drugs
ciency (HIV) virus infection (Navarro 2019). Ascendia and include the coated particles (membrane-diffusion
170 11 Formulation Development Concepts

mechanism) and molecularly dispersed systems (matrix- Baclofen GRS (a bronchodilator; Sun Pharma), which con-
erosion mechanism) (see Section 11.4). Ascendia nanotech- sists of a coated multilayer floating and swelling device; and
nology platforms may be found favorable in producing drug Glumetza (for blood glucose control; Depomed), which is a
products under section 505(b)(2) pathway (for information polymer-based system (Amin et al. 2008a). These units
on section 505, refer to Chapter 22), which allows compa- release the drug one layer at a time, starting with the most
nies to obtain FDA approval relying in part on clinical data superficial layer first. The outer layer in contact with aque-
or other companies’ literature (Huang 2019, 2020a). ous fluid swells and prevents further entry of the fluid
inside the matrix. When the outer layer swells, it entraps
some air and helps to keep the unit floating on the surface
of the fluid (Dubin 2007).
11.4 Rate-Controlled Drug Delivery Other systems contain matrices prepared from swellable
polymers and effervescent materials that upon contact with
Systems for controlling drug release from a dosage form the gastric fluid generate CO2 gas; the gas gets entrapped in
have been developed to maximize therapeutic benefits the polymer structure, helping the unit to float (Dubin
while minimizing adverse outcomes of drug therapy. Diffu- 2007). Once the outer layer is completely degraded and
sion systems and activation approaches are the two main the drug is released from it, a new outer layer is formed,
classes of rate-controlled drug delivery. Diffusion systems and the process continues until the entire unit is gone.
are of several types and include membrane permeation, The hydroxypropyl methylcellulose (HPMC), with appro-
matrix diffusion, and microreservoir dissolution. The acti- priate hydration rate and viscosity, used in these floatable
vation technologies utilize hydrodynamic, osmotic, or units is extremely popular in the preparation of con-
vapor pressure methods. Ion or pH activation, ultrasound, trolled-release systems. Its popularity stems from several
and magnetism are all activation delivery techniques features, including a high capacity for drug loading, easy
(Chien 1985). compression into tablets, having temperature- and
Membrane permeation allows a polymeric barrier to con- humidity-independent viscoelastic characteristics, being a
trol delivery of a drug housed inside a rigid compartment. crystallization inhibitor which helps enhance poorly solu-
Usually, the drug is in the form of dispersion, and its deliv- ble low-ionized APIs’ solubility and bioavailability (in par-
ery through the membrane follows a zero-order rate. Matrix ticular those containing a neutral amine group), and its
diffusion systems depend on dispersing the drug in a biode- inertness, among others (Gothoskar 2005; Stegemann
gradable matrix form. The rate of drug diffusion from the 2020). Floating foam-based systems are also used to provide
polymeric matrix is a function of the square root of time. gastroretentive drug delivery. The matrix of these systems
The microreservoir dissolution method employs a suspen- consists of porous (low density) material resembling foam
sion of the drug particles in an aqueous solution dispersed (e.g. Accurel MP; polypropylene foam powder). Drugs with
throughout a hydrophilic polymeric matrix. The suspen- or without added polymers are adsorbed on the foam par-
sion may be housed within an additional polymeric mem- ticles. The entrapped air within the matrix allows the unit
brane to further control the rate of drug release. The rate of to float upon exposure to an aqueous environment. The
drug release from the device is a function of its solubility in drug release profile from the system is independent of
the aqueous solution and in the polymeric matrix the foam materials; however, it depends on the type of pol-
(Chien 1985). ymer used with the drug (Amin et al. 2008a).
Some gastroretentive systems, for example, work via Other floating matrices utilize lipid materials as a base.
membrane permeation mechanisms. Floating drug delivery For example, lipid matrices known as Gélucires
systems that contain a drug dispersed in a hydrophilic (Gattefosse, St-Priest, France), composed of mixtures of
matrix (e.g. guar gum; hydroxypropyl methylcellulose) mono-, di-, and triglycerides with poly(ethylene glycol)
exhibit a density below than 1 when placed in an aqueous esters of fatty acids, have been shown to provide a sustain-
environment (such as gastric fluid) and thus float on the ing effect. Gélucires have hydrophile–lipophile balance
surface of the aqueous liquid (Jain et al. 2005). The hydro- (HLB) values in the range 1–18, with a wide melting-point
philic matrix can form a superporous hydrogel. Upon range between 33 and 65 C. Low-HLB-value Gélucires pro-
absorption of water, the hydrogel material swells with duce units with floating characteristics gastrointestinal
the formation of space within the matrix, known as the fluids. The absence or low content of PEG esters mixed with
effective pore size. When the effective pore size is larger than Gélucires provides a sustaining effect. [Larger amounts of
10 μm, the hydrogel is known as a superporous hydrogel PEG esters mixed with the lipid matrix accelerate the rate
(Amin et al. 2008a). Some examples of this technology of drug release (Amin 2008a, 2008b; Patel and Patel 2008).]
available on the market are Liquid Gaviscon (an antacid; Glyceryl monostearate retards the release of drugs from for-
GlaxoSmithKline), which is a floating alginate raft; mulations. When used in combination with Gélucires, the
11.4 Rate-Controlled Drug Delivery 171

matrix failed to float. Mixtures of ethylcellulose, a release An example of these activation technologies is the Oros sys-
retardant agent, with Gélucires provided both retardation tem (Alza Pharmaceuticals, Mountain View, CA; a subsid-
of drug release and floating characteristics. Gélucires com- iary of Johnson & Johnson). Oros drug delivery is based on
bined with HPMC showed an accelerated drug release pro- osmosis and has wide clinical applications, including drugs
file compared to Gélucires alone (Amin et al. 2008a). used in the treatment of attention-deficit hyperactivity dis-
Another oral delivery membrane permeation technology order, benign prostatic hyperplasia, hypertension, diabetes
is the Eudramode platform (Evonik Röhm GmbH, 2012, (type 2), overactive bladder, pain, and schizophrenia
Darmstadt, Germany), which is made of layered pellets. (Conley et al. 2006). Some drug products using Oros include
The core of the pellets is composed of modulator ions (a salt albuterol, chlorpheniramine maleate, doxazosin mesylate,
of weak or strong acids, such as sodium citrate) layered glipizide, ivermectin, nifedipine, and verapamil (Gupta
with a modulating film (Eudragit NE 30 D), followed by et al. 2005).
a layer of the drug, and a covering controlled-release layer Systems that depend on pH activation are covered by a
of quaternary ammonium ions (Eudragit RL/RS 30 D; mixture of polymers that deliver the device intact to the
ammoniomethacrylate copolymers). The pellets may be intestines. Under the pH conditions available in the intes-
placed in a capsule form or compressed into a tablet. The tine, some polymers are soluble and others are not. This
Eudramode platform was shown to deliver the drug in a causes the polymeric membrane surrounding the device
zero-order release fashion. Three factors play an important to become porous, allowing diffusion of the drug through
role in controlling the drug release from the pellets; these the pores. Ion activation systems rely on interaction of
are the salts used in the core and the thickness of both the drug with a resin, which upon delivery to the gastroin-
the modulating film and the controlled-release covering testinal tract undergoes reactions with available ions to
outer layer (Ravishankar et al. 2005). Eudragit polymers release the drug slowly from its resin–drug complex and
(Evonik Röhm GmbH, 2012, Darmstadt, Germany) are to provide controlled delivery (Chien 1985). Insulin encap-
copolymers derived from esters of acrylic and methacrylic sulated in nanoaggregate particles (100–230 nm) and later
acid. Their properties are based on functional groups coated with weak polyelectrolytes [(Poly(α, ß-1-malic acid)
located on the polymeric chain. Eudragit polymers are and chitosan] exhibited a pH-dependent release of insulin
available in a wide range of applications, including aqueous from the nanoparticles due to the sensitivity of the polyelec-
dispersions (L50 D-55; NE 30 D; NE 40 D; NM 30 D; RS trolytes to pH. At physiological blood pH (7.4), insulin
30 D; RL 30 D; FS 30 D), powders and granules (L100; release from the matrix was observed, whereas at pH 4–5
L100-55; S 100; RS 100; RL 100; RS PO; RL PO; E-100; no insulin release was detected. Furthermore, the addition
E PO), and organic solutions (E12,5; L12,5; S12,5; RS12,5; of a higher number of polyelectrolyte layers to the surface of
RL12,5) (Evonik Röhm GmbH). Eudragit polymers can the nanoaggregates resulted in a reduction of the rate of
be classified into two main categories: pH-dependent and release of insulin from the nanoparticles (Fan et al.
pH-independent. As pH-dependent films, the polymers 2006). In addition to the pH, drug lipophilicity as measured
become soluble in solution if the pH exceeds a certain by the partition coefficient (log P) was shown to play an
value, whereas pH-independent polymer films act as a bar- important role in drug release (Sawant et al. 2010). For lido-
rier, allowing diffusion of a drug through the film. When caine (log P = 2.6; lipophilic) and lidocaine hydrochloride
used as a matrix, the polymer acts as a sponge, releasing (log P = 0; hydrophilic), it was observed that when lido-
the drug in a controlled fashion. The drug diffuses out of caine was incorporated into a hydrophilic matrix (hydroxy-
the pH-independent matrix, whereas it is released by ero- propyl cellulose), the release rate of the drug followed a
sion and diffusion when a pH-dependent matrix is used slow non-Fickian release rate, whereas the hydrophilic salt
(Álvarez et al. 2007). of the drug experienced a burst effect for its release from the
Activation technologies that control drug delivery by a matrix. When lidocaine was incorporated into a hydropho-
pressure mechanism depend on interaction between the bic carrier (Eudragit E100), a burst effect was noted. It
device and its environment following administration. Usu- appears that lidocaine interacted with hydroxypropyl cellu-
ally, the drug is housed within a physical compartment, and lose, which resulted in a slow release of the drug from the
its rate of release through an orifice is controlled by either matrix. No interaction between Eudragit E100 and lido-
driving water inside the device through osmosis, absorbing caine was noted, which resulted in a burst effect (Sawant
water by a polymeric matrix that upon swelling exerts pres- et al. 2010).
sure on the drug reservoir or by the use of a chemical that Activation systems that depend on magnetism employ
vaporizes at body temperature, with the gas thus formed pellets carrying a tiny magnet within them. The drug–pellet
exerting pressure to release drug from its compartment. system is coated with impermeable polymeric membrane to
These systems allow release of the drug from the device the drug, except at the center of the pellet, where no coat is
by a zero-order process (Chien 1985; Gupta et al. 2005). applied. The drug is released from the uncoated center area,
172 11 Formulation Development Concepts

and its rate of release is accelerated using an external mag- stage of dissolution and absorption occurs in the gastroin-
netic field (Chien 1985). Applications of magnet-containing testinal (GI) tract after the patient swallows the resulting
units may be helpful in situations where an increase in the “suspension” made of the disintegrated particles
drug release rate is desirable at a certain point in time. For (OraSolv, CIMA Labs, Inc., 2012, MN). ODT technology
example, insulin delivery through such a device would be can also offer a sustained-release delivery mechanism such
desirable, where the patient can activate delivery of insulin as the tablets developed by Eurand N.V. (2012) (AdvaTab,
when needed after meals (Langer 1985). Another possible Amsterdam, The Netherlands). AdvaTab tablets dissolve
external activation energy source is ultrasonic waves. quickly within the mouth cavity (15–30 seconds) and have
A biodegradable polymeric matrix containing the drug the capacity to carry large doses of drug (Eurand N.V. 2012).
may be used, and the rate of polymer degradation is In general, an ideal ODT unit should dissolve rapidly in a
enhanced by an ultrasonic device, thus facilitating release matter of seconds within the mouth cavity and should have
of the drug from its matrix (Chien 1985). a taste-masking property and a smooth mouth feel
(Hamilton and Lutz 2005). In addition to tablets, oral
fast-dissolving films or strips (OFDSs) are also available to
11.5 Drug Delivery Technologies enhance the delivery of poorly water-soluble and permea-
for Improving Oral Delivery ble APIs. The film is composed of cellulose-based polymers
or poly(ethylene oxide) containing strong flavoring agents
The oral route of administration has been by far the most to mask the taste of added ingredients. Examples of these
convenient method of drug administration. Over 50% of oral strips are Triaminic, Chloraseptic, and Listerine (Ali
the drugs available on the market exist in this form (Jain and Quadir 2007). The normal size of an OFDS is about
et al. 2005). The ease of delivery combined with a relatively the dimension of a postage stamp. Upon contact with the
rapid onset of action, along with lower cost per dose unit, saliva, the film disintegrates instantaneously. Original for-
makes this route an ideal way for augmenting the therapeu- mulations of OFDSs were introduced on the marker as
tic effects of drugs. One of the recent innovations in enhan- breath-freshening films. These original breath-fresheners
cing the delivery of drugs via this route is the use of orally were followed by introducing products for delivering vita-
disintegrating tablets (ODTs). While oral delivery of drugs mins, minerals, and dietary supplements. The advantages
by conventional dosage forms (tablets, capsules, etc.) of OFDSs are fast disintegration and dissolution, flexibility
requires the intake of water to facilitate swallowing the of the film, and nonfriability (Hariharan and Bogue 2009).
solid dosage form, the rapid disintegration and dissolution The permeation of drugs through the GI tract after an
of ODTs in the mouth cavity eliminate the need of water oral administration is crucial for delivering a sufficient
during administration. Examples of this dosage form are amount of the drug to the circulation in a timely manner.
Ondansetron ODF/Setofilm® (Lavasta Pharma FZ-LLC, This becomes even more important if the drug has a poor
Dubai, UAE; for nausea) and Zolmitriptan Rapidfilm® permeability profile through the gut mucosa. A system
(Applied Pharma Research/Labtec, Switzerland/Germany; developed by Merrion Pharmaceuticals (2012), known as
for migraine headache). Characteristics that are considered GIPET (gastrointestinal permeation enhancement technol-
to be highly desirable for ODT’s bulk excipients are (Joshi ogy), facilitates and enhances the absorption of poorly per-
and Duriez 2004) (1) delivers good dispersion and fast dis- meable drugs up to 200-fold. GIPET works via a proprietary
solution (in seconds) upon administration, (2) provides an micelle–drug membrane transport enhancement method.
agreeable mouthfeel while masking the bad taste of drugs, This technology is applicable to a wide range of drugs,
(3) remains stable under various storage environmental including biotechnology products. Because of its good
conditions (e.g. temperature, humidity, etc.), (4) allows ade- safety profile, the FDA has included GIPET in its GRAS
quate and sufficient loading of the drug within its matrix, (generally regarded as safe) listing. Nanosplode, Lip’ral,
and (5) resists the shocks/stresses imposed on it from Lip’ral Synchronized Solubilizer Release, and Hydroance
patient’s handling and during transportation. One ODT (Lipocine, Inc., 2012, Salt Lake City, UT) are other technol-
technology uses a specialized lyophilization method (i.e. ogies that facilitate the absorption of drugs that suffer from
Zydis, Catalent, New Jersey) in which the entire formula- poor absorption and/or a poor solubility profile from the GI
tion process is prepared within a blister compartment. tract. [It is estimated that about 40% of all drugs have poor
The Zydis fast-dissolving technology (within three seconds solubility in water (Dubin 2005a; Chattopadhyay and She-
when placed in an aqueous environment) has been shown kunov 2006).] Following rapid disintegration from solid
to improve the bioavailability of drugs, as well as the onset dosage form (e.g. capsules), the drug molecules are
of action, comparable to that of an intravenous injection engulfed with specialized proprietary lipids that facilitate
(Catalent Zydis brochure 2007). ODTs prepared by a direct drug absorption by the GI mucosa (Lipocine, Inc. 2012).
compression method produce tablets that disintegrate and Another important area for delivering drugs is that to the
dissolve rapidly in the mouth cavity; however, the final brain and the central nervous system (CNS), which is often
11.7 Drug Delivery Technologies for Transdermal Delivery 173

limited due to the presence of the blood–brain barrier mucoadhesive property of tablets using a rat stomach mem-
(BBB). The BBB is due to the presence of tight junctions brane model. Of all the single agents tested, HPMC had the
in the blood vessels of the CNS that do not permit drugs best mucoadhesive strength. When other agents were
to pass through them, natural protective measure for the mixed with HPMC, a synergistic mucoadhesive effect was
brain. LipoBridge (Genzyme Pharmaceuticals 2012, Massa- seen. A combination of chitosan: carbopol (1 : 4) gave the
chusetts) is a short-chain oligoglycerolipid compound that highest strength for mucoadhesion. Overall, it appears that
reversibly opens the tight junctions, thus facilitating the combination polymeric mixtures provide a stronger
entry of drug molecules into the CNS and brain tissues. This mucoadhesive force than that of a single agent alone.
allows an effective concentration of drugs in the CNS and
brain to increase by a factor of up of 100 above normal.
The system appears to be safe and is excreted unchanged
without being metabolized (Genzyme Pharmaceuti- 11.7 Drug Delivery Technologies
cals 2012). for Transdermal Delivery

The skin is the largest organ of the body. It presents a formi-


11.6 Drug Delivery Technologies dable barrier for foreign substances to enter the body and for
for Improving Transmucosal Delivery endogenous ones to escape to the outside. The presence of ker-
atin in the stratum corneum prevents water from entering or
Transmucosal delivery is seen as somewhat of an extension leaving the skin (except at sweat pores). For normal skin, the
to other routes. Via this system, drugs are transported pH is slightly acidic (pH 5–6), due to the presence of organic
through a transmucosal pathway by placing them in the acids on the surface of the skin, such as amino acids and fatty
mouth, nose, or another route. The market for transmuco- acids. Transdermal delivery of the drug can be activated by
sal delivery is estimated to be in billions of US dollars annu- various physical mechanisms including iontophoresis, sono-
ally. The transport of drugs through the mucosa of the phoresis, electroporation techniques, Magnetophoresis, and
mouth cavity is reported to be highly efficient, exceeding micro-needles (Lopes and Soares 2015; Sirisha and Sailaja
that of the skin manyfold (4–4000 times higher). An exam- 2018) (see Chapter 19). In general, transdermal drug diffusion
ple of such a delivery system is the OraVescent technology may be enhanced by a reduction in the diffusivity resistance
(CIMA Labs, 2012, Brooklyn Park, MN), which offers through (1) dissolution of drug in its vehicle; higher concen-
mucosal transport of drugs from the mouth cavity (sublin- tration leads to higher flux of the drug through the stratum
gually or between the buccal and gingival tissues). OraVes- corneum, (2) disruption of the lipid structure of stratum cor-
cent is available in compressed tablet form and is dispensed neum, which permits local diffusion enhancement of drug
in a PakSolv proprietary blister package (Brown and molecules, (3) interaction with intercellular proteins, and
Hamed 2007). Other advantages of the buccal delivery over (4) improving drug partitioning into the stratum corneum,
the oral route are avoidance of the gastrointestinal degrada- which facilitates the diffusion of drug from the vehicle to
tion and the first-pass effect as well as the ability to deliver the stratum corneum (Sirisha and Sailaja 2018).
the drug in patients who have difficulty swallowing Since the first transdermal product appeared on the US
(Davidson and Rousset 2018). market in 1979 (Scopolamine for the control of nausea
Sodium alginate is another type of material found useful and vomiting), the transdermal delivery applications of
through its bioadhesive properties, thus allowing delivery drug products have gained interest in improving patient
of APIs through the GI tract and nasal mucosa. Sodium compliance, avoiding hepatic first pass metabolism, and
alginate is a biodegradable natural polysaccharide that reducing the occurrence of gastrointestinal side effects of
forms viscous solutions upon dissolution in water drugs. Systemic side effects are also reduced by providing
(20–400 cP). Its viscosity is pH-dependent, and it decreases controlled release of the drug while enabling a steady blood
at pH values above 10. It can be used as a matrix in floating drug-level profile (Sirisha and Sailaja 2018).
beads and tablets of gastroretentive preparations. Research Transdermal patches are often used to deliver drugs
on sodium alginate has also revealed its usefulness to pro- through the skin. The solubility of drugs in the adhesive
vide or enhance the controlled-release delivery of drugs, as material of the patch can be difficult to estimate. Myatt
well as delivering proteins, DNA, and various other carriers et al. (2008) presented a practical method for estimating
(e.g. liposomes and microspheres) (Patel et al. 2007). With the solubility of solutes in the adhesive matrix. In this
respect to the mucoadhesive properties, various agents may method, the amount of drug diffused over time is obtained
be used to provide such action. Among these agents are by diffusion experiments, and the data are then plotted to fit
chitosan (cationic), carbopol 934 (anionic), and hydroxy- Higuchi’s model:
propyl methylcellulose (HPMC, nonionic). Majithiya
et al. (2008) studied the effect of these agents on the M = k0 t0 5 11 1
174 11 Formulation Development Concepts

where M is the cumulative amount of drug released per unit companies must consider three critical and essential areas:
area, t is the time, and k0 is Higuchi’s constant. The value of (1) safety, (2) purity, and (3) consistency, with the manufac-
k0 is calculated at different starting concentrations of the turing processes being scalable and reproducible (Washer
solute added to the adhesive matrix. When k0 values are and Knop 2020). During pandemics, the pressure exerted
plotted against the initial concentration of the drug in the on biotechnology companies to produce billions of doses
matrix, the graph forms two distinct linear portions that rather than millions fast becomes a reality. Companies have
intersect at a point. The point of intersection corresponds to re-invent operation areas such as quality control to meet
to the solubility of the drug in the adhesive matrix read the global demand without compromising safety while
directly from the x-axis (Myatt et al. 2008). maintaining speed, capacity, and product quality
(Anonymous 2020b). During the COVID-19 pandemic,
the CRISPR technology was implemented to develop
FDA-approved rapid-testing platforms for the SARS-CoV-
11.8 Special Considerations for 2 virus (Straiton 2020).
Biotechnology and Protein Delivery The future of pharmaceuticals will include a great num-
Systems ber of biotechnology products that are composed of pro-
teins. Sterility, purity, and stability (physical and
The biotechnology industry has two main components: chemical) are the obvious challenges in this area of deliv-
“upstream” processes and other related to “downstream” ery. Each area should be addressed independently since a
processes. The upstream elements deal with materials stable product does not guarantee sterility or purity, and
and issues related to producing the product, while down- vice versa. Considerations for storage conditions (tempera-
stream processes handle the product’s purification to its ture, cleanliness) and handling (shaking the container vs.
final active form. Incidentally, downstream processing swirling) are also areas of concern for these delivery systems.
comprises 80% of the total cost associated with generating It is known in pharmacy practice that vigorous shaking of a
biotherapeutic agents (Endres and Dabre 2020). Biotech- protein product produces breakage in the chain of the pro-
nology’s birth is believed to have occurred with the inven- tein, rendering it denatured (Chan 2008). For biotechnology
tion of recombinant DNA by Stanley N. Cohen and Herbert products, their solutions may be filtered to remove any living
Boyer in 1973 (Shaffer 2020). The biotechnology applica- or nonliving organisms from the product. A 0.22 μm pore fil-
tions are numerous and include immunotherapy, ter can be employed to remove bacteria, and nanofilters are
CRISPR-Cas9 gene editing (CRISPR stands for Clustered utilized to remove viruses from the solution. When it comes
Regularly Interspaced Short Palindromic Repeats, and to biotechnology agents and products, the term “biosimilar”
Cas9 is an RNA-guided endonuclease found in certain bac- is used in place of “generic drug” due to their large molecular
teria), and gene therapy (Shaffer 2020). CRISPR was ini- structure, and the different manufacturing processes used
tially discovered by Japanese scientists in bacteria during that do not assure consistency in their “identity, purity,
the late 1980s and by another scientist from Spain in the potency, and safety” (see Chapter 24) (Aziz 2020).
1990s while working on genetic material sequencing in Another area of concern is that proteins in formulation
Archaea (single-celled prokaryotic organisms whose cells often aggregate in the form of dimers to multimers. The
exhibit undefined nucleus) (Behlke 2020). These repeats dimers are usually soluble in pharmaceutical solvent sys-
in the bacterial genetic code served as a defense mechanism tems (generally, aqueous in nature), while larger aggregates
for the bacteria to protect itself against subsequent viral may grow to sizes that become visible to the naked eye.
infections (i.e. bacterial immunity against viral infections) Aggregate formation is believed to give rise to immunoge-
(Behlke 2020). Emmanuelle Charpentier and Jennifer nicity, as well as to create major challenges in the areas of
Doudna were awarded the prestigious Nobel Prize in chem- handling, production yield, and quality (Das and Nema
istry in 2020 for making the genetic editing machinery pos- 2008). Detection of particulate matter due to aggregation
sible in vitro using the Cas9 enzyme system (obtained from of protein molecules can be detected by various measures,
Streptococcus pyogenes) as a platform and applying it to including laser diffraction particle analyzers, dynamic
mammalian cells (Jinek et al. 2012). Their work has poten- image analysis, imaging, and Raman spectroscopy (Das
tial applications in a variety of areas such as agriculture and and Nema 2008). Moreover, the pharmaceutical industry
medicine. Gene therapy employs, for the most part, viral utilizes various techniques for testing the structural integ-
vectors (70% of the cases) to deliver genes to the host cells rity of proteins during processing. These include capillary
(Lingelbach 2020). The types of viral vectors used include electrophoresis, high-performance liquid chromatography
adenoviral, lentiviral, and adeno-associated viruses methods, mass spectrometry, glycosylation analysis, amino
(Lingelbach 2020). Concerning the manufacturing of acid analysis and primary sequencing by Edman chemistry,
viral-vectored gene therapy products, biotechnology spectroscopy techniques, differential scanning calorimetry,
11.8 Special Considerations for Biotechnology and Protein Delivery Systems 175

dynamic and static light-scattering methods, and analytical and protein structure can not only improve solubility but
ultracentrifugation/sedimentation techniques, among can minimize aggregation and adsorption of the proteins
others (Chan 2008). Raman spectroscopy and Raman as well (Dubin 2008).
microscopy are commonly used to monitor the stability New approaches, such as Emisphere’s Eligen (2012,
of proteins during processing, allowing in situ testing, as Cedar Knolls, NJ), are promising technologies for oral
well as testing of samples in a rapid manner by matching delivery of drugs that are otherwise limited to the paren-
Raman fingerprinting of proteins with that of samples. teral route. This technology has been tested with many
However, Raman technology does not work well with lyo- important drugs, including calcitonin, growth hormone,
philized material or when the concentration of protein is heparin, and insulin (Majuru 2005). It utilizes a proprietary
too low (Wen et al. 2010). delivery agent that when given along with a drug orally
Routes of administration for biotechnology products are forms a complex in the GI tract, which changes the physical
also limited to the parenteral route, although pulmonary characteristics of the drug, allowing it to be absorbed
and nasal delivery may be alternative options. For example, through the mucosa. Following absorption, the complex,
the delivery of insulin is done primarily by syringes, pens, which is held together by weak intermolecular forces, dis-
jet injectors, and pumps (Shankar 2005) and attempts to sociates, releasing the active drug to exert its pharmacolog-
market insulin as a buccal spray or in inhalers have been ical effect.
seen recently. Generex Biotechnology is already marketing Emisphere’s Eligen technology has been placed on hold
insulin as a buccal delivery device (Oral-lyn), which is by the company with respect to developing it further as a
approved for use in several countries (Amin et al. 2008b). means of delivering insulin orally. In a statement on its
Inhaled insulin preparations have been introduced to the website, the company declared that “Oral insulin could
market, but for varying reasons (mainly safety issues, effi- represent a significant opportunity for Emisphere and, in
cacy, and cost) they were withdrawn by the manufacturer theory, would meet unmet market needs. However, realis-
within months of being offered (e.g. Exubera, Pfizer). Exu- tically, at this stage in its development and based on what
bera was a fine powder (particle size 1–5 μm) of bioavailable has been and perhaps what has not been done with this
human insulin. Novo Nordisk’s AERx Insulin Diabetes product candidate, it presents significant challenges. At this
Management System was halted in its development stage time, the Company is exploring its strategic options for the
before reaching the market. This system was an inhaled oral insulin compound.” This illustrates the types of chal-
insulin system in liquid form. Eli Lilly, in cooperation with lenges facing oral delivery of peptides and proteins.
Alkermes, has been developing powdered insulin for inha- Delivery of insulin via other routes, besides the paren-
lation that could be delivered using a simple inhaler. The teral route, has been investigated fiercely for many decades.
particles are large and have low bulk density, providing The main reason for this interest is its importance in man-
them with good aerodynamic properties. Other devices aging diabetes, a major health issue worldwide. Diabetic
for inhaled insulin are also being developed by several com- patients are required to self-inject insulin preparations sev-
panies (Baxter BioPharma Solutions, BioSante Pharmaceu- eral times a day, a painful and inconvenient way to main-
ticals, Kos Pharmaceuticals, and MannKind) (Amin tain a normal lifestyle. Thus, developing alternative routes
et al. 2008b). of administration has been a priority in this field. The oral
Recent work in the area of oral delivery for peptides and route, in particular, is of greatest interest, as it provides a
proteins is also showing some hopeful signs of progress, way that mimics the body’s own method of insulin “deliv-
although issues related to the reproducibility of delivery ery.” Insulin is a high-molecular-mass hormone of 5800 Da.
remain a big obstacle. The presence of food in the GI tract, It is synthesized by the beta cells in the islets of Langerhans
the availability of active enzymatic processes, and the chal- of the pancreas and transported via the portal vein to the
lenges facing absorption of high-molecular-mass molecules liver. Similar transport of insulin to the liver occurs via
by the gut mucosa are just a few factors that render oral the oral route, which is the natural way for the body to han-
delivery of proteins and peptides the “holy grail” of drug dle insulin. Unfortunately, the GI tract is an extremely hos-
delivery. Another area of concern is that related to the sol- tile environment for insulin, due to the presence of
ubility of API after being released at the site of administra- enzymes capable of destroying this regulating hormone.
tion. Improvement in the solubility of peptides and proteins The main enzymes are pepsin, trypsin, and chymotrypsin.
may be facilitated by the inclusion of counterions in the for- Studies performed in vitro have shown that the effect of
mulation. Acetate and trifluoroacetic acid (TFA) are com- these three enzymes is rapid (seconds to minutes) and
monly used as counterions in protein formulations. highly effective (80–100% degradation). Pepsin requires
Higher residual TFA levels are of concern; however, they an acidic environment to act, whereas trypsin and chymo-
are tightly regulated because they can cause undesirable trypsin need a slightly basic pH. This pH requirement cor-
effects in patients. Also, modifications made to peptide responds to their location in the gut cavity; pepsin is located
176 11 Formulation Development Concepts

in the stomach, and the other two enzymes are found in the addition of an aqueous solution (w2) containing 2%
small intestine. Another obstacle for the oral delivery of poly(vinyl alcohol) (PVAL). The resulting double emulsion
peptides and proteins is the intestinal permeability. By is mixed with a PVAL aqueous phase (0.3%), and the
far the most important area for drug absorption is the duo- organic phase is evaporated to form a suspension of nano-
denum. However, the absorbing epithelium is limited in its particles. An example of these microparticles is Novartis’s
action to smaller-molecular-mass drugs and more lipo- Sandostatin LAR Depot (composed of a biodegradable glu-
philic solutes. As such, peptides and proteins are not good cose star polymer with a copolymer of lactic and glycolic
candidates for absorption in this region, and thus they have acids) delivering the chemotherapeutic agent octreotide
a very low oral bioavailability (Cheng and Lim 2008). acetate, used in the treatment of acromegaly (Bilati
Recently, the French-based company Gattefossé has intro- et al. 2005).
duced self-emulsifying drug delivery systems (SEDDS) that Delivery of peptides and proteins through the pulmonary
work by solubilizing the peptides in the gastrointestinal route is associated with significant challenges regarding the
tract while protecting them from degradation by digestive stability of proteins during transportation, use, and storage.
enzymes. The SEDDS have been promising applications Metered-dose inhalers (MDIs) contain propellants that are
for oral delivery of peptides such as desmopressin, insulin, not compatible with most proteins. Thus, nebulizer solu-
and leuprorelin. SEDDS are also employed to enhance the tions containing unbuffered isotonic solutions with a pH
aqueous solubility of poorly soluble compounds, which in of 5 or higher are preferred over MDIs for delivering protein
turn render them suitable for oral administration. Solid drugs. Calcium salts (calcium chloride) are often used in
SEDDS may also be applicable for processes such as hot protein solutions as stabilizers. Surfactants such as Tween
melt extrusion and granulation. 80 are usually added to protein solutions to minimize the
Pharmaceutical companies commonly rely on the concept adherence of protein to the container walls and to protect
of the human maximum absorbable dose (MADhuman) as a the protein from aggregation and a rise in temperature dur-
measure of drug bioavailability. MADhuman measures the ing nebulization. Instead of solutions, powdered aerosols
oral dose absorbed (mg) (Dabs) that reaches the circulation may be used where the protein is present in freeze-dried
as compared to the oral dose administered (Timpe 2007): form. To facilitate the flow of the protein’s powder, an inert
coarse powder carrier is usually mixed with the protein par-
Dabs = P S A T 11 2
ticles. Protein products should be protected from high tem-
where P is the apparent permeability coefficient (cm/s), S peratures and humidity during storage, as these conditions
the saturation solubility of an API (mg/mL), A the surface lead to denaturing the protein through aggregation reac-
area available for absorption (cm2), and T the transit time tions. The bioavailability of protein drugs via the lungs
through the small intestine (seconds). In humans, the esti- can be restricted, as proteolytic enzymes and the presence
mated values for A and T are 800 cm2 and 3.3 hours, respec- of macrophages act to eliminate the protein and limit its
tively. The Dabs desired is often stated as being 40–60% of absorption (Krishnamurthy 1999).
the efficacious dose (ED50) (Timpe 2007). It is expected that Other areas of biotechnology products are related to gene
the value of Dabs with respect to the oral absorption for delivery. Liposomes are one of the preferred carriers for
unaided peptides and proteins is very low or nearly zero. genes, due to their potential low immunogenic reaction
A promising area for delivering proteins orally is the (Felgner et al. 1987). In general, liposomes are classified
entrapment of the API in a polymeric mesh, from which into three types: multilamellar vesicles (MLVs), small unila-
the drug is released slowly overtime, as well as being pro- mellar vesicles (SUVs), and large unilamellar vesicles
tected from enzymatic degradation in the gut. An example (LUVs). With respect to size, MLVs range between 0.1
of a system that has produced positive experimental results and 5.0 μm, whereas SUVs and LUVs are 0.02–0.05 μm
in this respect is the polymer system known collectively as a and greater than 0.6 μm in size, respectively. There are
thiomer. These polymers are essentially thiolated and pro- two major methods of producing liposomes: by the forma-
vide a zero-order drug release kinetic from their matrix. The tion of emulsions and by hydrating a dried lipid film fol-
matrix itself is made of a three-dimensional structure that lowed by vigorous agitation (sonication or high-shear
offers a tightened tablet matrix for the delivery of peptides impeller) (Chrai et al. 2002). As a carrier for genes, lipo-
and proteins (Langoth and Bernkop-Schnürch 2005). somes were first suggested by Felgner et al. (1987). The abil-
One area for protein delivery employs an encapsulated ity of liposomes to travel through cytoplasmic and nuclear
ion of proteins in biodegradable micro- or nanoparticles. membranes is essential for delivering genes to the DNA. In
The production of these particles involves a double- addition, genes thus delivered should be able to integrate
emulsion method, in which the aqueous phase containing with the cell’s DNA to become effective. Cationic liposomes
proteins (w1) is emulsified with the polymeric material dis- are commonly used to deliver the plasmid gene, because
persed in an organic phase to form aw1/o emulsion. these liposomes were found to be most stable in vivo
A double emulsion (w1/o/w2) is then formed by the (Bellare and Daga 2005).
11.9 Drug–Excipient and Excipient–Excipient Interactions 177

Generally speaking, proteins are prepared as lyophilized the excipient’s safety before use (Dubin 2005a; Eatmon
dried powders, in solutions, or as frozen preparations. The and Loxley 2009). The approved list for excipients for cer-
latter has its own advantages and disadvantages. Frozen tain formulations may be even more limited, such as those
protein preparations are desirable during batch manufac- used in parenteral preparations (Dubin 2005a). Generally
turing of a drug product. They reduce the waste associated speaking, pharmaceutical companies opt to use an excipi-
with the manufacture of these products, as well as assuring ent blend chosen from an FDA-approved excipients list.
that the end product meets its specifications during storage Characteristics of a good excipient are compatibility with
and throughout its shelf life. In addition, the low freezing the API and other added ingredients in a formulation, being
temperature (−20 C) affords a hostile environment for a nontoxic substance, and its availability on the market
microorganisms and thus helps in contamination control with reasonable purity and an acceptable grade (Eatmon
of the product. However, freezing and thawing cycles can and Loxley 2009). Some of the excipients used in biophar-
cause denaturation of protein. Also, solution heterogeneity, maceuticals may require specific handling. For example,
commonly found in the frozen material, can result in phys- human serum albumin (HSA; 66,000 Da) is often used as
ical and chemical alterations that have a detrimental effect an excipient in biotechnology formulations. One of require-
on a proteineous drug (Kantor et al. 2011). ments of its use is that it be free of any viral contamination.
This is accomplished by heating HSA to 60 C for 10 hours.
This excipient has the advantage of being relatively stable at
room or higher temperatures, safe to use due to lack of
11.9 Drug–Excipient and immunogenicity, and can be used to eliminate the adsorp-
Excipient–Excipient Interactions tion of other proteins from the surface of containers. HSA
is also available from recombinant DNA technology
The market for pharmaceutical excipients is expanding at (Alburex, Mitsubushi Welfarma, Japan), which eliminates
an accelerated rate and is expected to reach $8.53 billion the problem of virus contamination (Chaubal 2005).
by 2023 (it was $6.4 billion in 2018) (Dubin 2019). The Other considerations in drug development are related to
choice of excipients in a formulation is not a marginal how excipients and drugs interact with each other in the
matter. Excipients may contain undesirable contaminants same formulation. These interactions may affect the phys-
that need to be identified prior to incorporating them in ical characteristics of final products, such as the dissolu-
formulations. Guidelines for impurities of excipients are tion and disintegration time for tablets. For example, an
traditionally listed in various pharmacopeias. For example, increase in the mixing time after adding magnesium stea-
the United States Pharmacopeia monograph for rate (a lubricant) to a mixture of ketorolac tromethamine
poly(ethylene glycol) lists the limits for major and minor with spray-dried lactose, cornstarch, pregelatinized
contaminants such as ethylene glycol (0.25%) and ethylene starch, or a combination, resulted in a decrease in the dis-
oxide (10 ppm) (Erickson 2005). The functions of excipients solution rate of the capsules containing the formulation
in formulations are numerous and include roles as solubi- with cornstarch but not with that containing pregelati-
lizing agents, stability enhancers, tonicity-adjusting compo- nized starch. The disintegration time of the capsules with
nents, preservatives, and release-controlling materials, cornstarch increased with an increase in mixing time with
among others (Dubin 2005a). Interactions among excipi- magnesium stearate but not with pregelatinized starch.
ents in the same formulation or between excipients and These effects on dissolution time and disintegration time
an API can lead to altered or diminished bioavailability were explained by the interaction between cornstarch
and/or product stability (Dubin 2005b). Isothermal stress agglomerates, magnesium stearate flakes, and drug parti-
methods are often used to test for incompatibilities existing cles, as documented by scanning electron microscopy. No
among ingredients in the same formulation. High tempera- such interactions were seen with pregelatinized starch
tures with or without moisture are used as storage condi- (Chowhan and Chi 1985a). When crospovidone was used
tions to determine how the various ingredients may in the formula, replacing cornstarch and pregelatinized
affect the stability of an API in a formulation. However, starch, similar effects were observed on the dissolution
the use of high temperatures during stress testing has its and disintegration times. Scanning electron microscopy
limitations if the ingredients melt at elevated temperatures revealed that drug particles get entrapped in crospovi-
(e.g. lipid components). For these formulations, extrapola- done, and the resulting agglomerates interact with
tion from high temperatures to ambient temperatures may magnesium stearate flakes as mixing time increased. Lar-
be problematic (Holm 2010). Most pharmaceutical compa- ger drug particles had more efficient entrapment with
nies that operate on a limited tight budget choose already crospovidone particles, resulting in a more pronounced
acceptable excipient blends by the regulatory agencies for effect on dissolution and disintegration time (Chowhan
their formulations. Otherwise, the company has to justify and Chi 1985b).
178 11 Formulation Development Concepts

11.10 The Presence of Contaminants particles from being present in the products released to
in a Formulation the public. It is estimated that about 50% of the recalls
issued in sterile parenteral products during the period
Chemicals used in formulations are available in various between 2010 and 2017 were due to this type of concern
purity grades. The pharmaceutical industry utilizes those (Laskina 2020). Parenteral vials have closures to protect
with characteristics that match those approved by regula- the product from contamination upon storage and hand-
tory health agencies. In particular, preparations that are ling. The United States Pharmacopeia addresses parenteral
intended for parenteral administration must contain che- container-closure integrity with guidelines found in chap-
micals of the highest possible purity available on the mar- ter 1207 (USP <1207>). Other forms of contaminants in
ket, with no or an acceptably low content of contaminants parenteral sterile products are leachables and extractables.
as determined by the health authorities. In certain cases, With increased use of newly synthesized polymers [poly-
such as the formulation of sterile single-use liquid biophar- ethylene, polypropylene, polyester-laminated polymers,
maceuticals, the development process requires the use of and poly(vinyl chloride)] as container materials for phar-
stainless steel bulk storage tanks for an API that contains maceutical and biological products, leachables and extrac-
several elements, such as aluminum, carbon, chromium, tables have become of great concern to the pharmaceutical
cobalt, copper, iron, manganese, molybdenum, titanium, industry (Northup 2008). This is true not only for sterile
tungsten, and zirconium. These elements may leach from injectables but also for preparations intended for aerosol
storage tanks and contaminate the product during the inhalations, ophthalmic products, and nasal aerosol sprays
development process (Leveen 2010). Analytical methods (Northup 2008). Regulatory agencies have published guid-
for detecting impurities in APIs or starting materials are ing principles to suggest that the best way to test for leach-
based on various chromatography techniques such as ables is with the actual formulation (Scott et al. 2020).
high-performance liquid chromatography (HPLC) and Examples of leachables in orally inhaled and nasal drug pro-
supercritical fluid chromatography. The latter method ducts include the polycyclic aromatic hydrocarbons (PAHs),
was found to be much superior over HPLC techniques in n-nitrosamines, and 2-mercaptobenzothiazole (2-MBT). All
detecting impurities in APIs and other starting additives of these chemicals are considered to be carcinogens. PAHs
(Zelesky 2008). and n-nitrosamines appear frequently in the environment,
Particulate matter contamination of sterile products is including in food products, soil, water, and air. 2-MBT resi-
another area of concern in the pharmaceutical industry. dues are found in rubber products and are known to cause
Particulates can enter sterile products during processing, allergic reactions in some people who wear rubber gloves
packaging, or use. Glass, plastic, and rubber materials that (Norwood and Mullis 2009). In general, leachables are
are used in vials and ampoules could contribute particulate chemical substances (e.g. zinc ions) that migrate from a con-
matter to the products housed within them. Other sources tainer’s surface into a product concentrate during normal
of particulates include personnel entering and working in storage and handling conditions; whereas extractables are
sterile packaging areas, sealing operation for ampoules, chemical substances (e.g. carbon-based compounds) that
and gas purging of containers without a filtration step. It can be obtained from a container’s surface after treatment
is always easier to prevent contamination from occurring with solvents under varying conditions of temperature
than to clean a contaminated product. Accordingly, com- and time (Castner et al. 2004). For example, in April 2010,
bining multiple steps during manufacturing, such as filling, Johnson & Johnson recalled its children’s formulations, cit-
sealing, and printing (in that order) under one-time opera- ing the presence of the pesticide and flame retardant 2,4,6-
tion may minimize the risk of introducing particulates into tribromophenol in these preparations. This pesticide, which
the product (Dean 1985). Wormuth et al. (2019) conducted originated from the shipping pellets, had permeated the
a study on the presence of particulate matter in biopharma- packaging walls, including the actual container of the prod-
ceuticals with single-use systems. Their findings indicated uct, and reached the formulation (Lynch 2011). Recommen-
that all bag products tested contained particles within the dations to overcome the presence of leachables and
visible range. The authors advised that the manufacturer extractables are to obtain information on the raw materials
should implement tightened controls to reduce the level and equipment used in making the containers, to utilize var-
of particulate matter present to an acceptable level. The ious methodologies in their identification, and to be aware of
United States, European, and Japanese pharmacopeias their sources, which may be from outside the container itself
require that the final parenteral product is free from visible (Castner et al. 2004). Various analytical techniques can be
particles. Parenteral manufacturers do not release their utilized in the detection of leachables and extractables.
batches until 100% of the units produced are visually The list includes single-crystal x-ray spectrometry, Fourier
inspected for particulate matter and other defects transform infrared spectrophotometry, diode-array ultravi-
(Laskina 2020). However, that does not preclude visible olet spectrophotometry, nuclear magnetic resonance
References 179

spectroscopy, and mass spectrometry. The latter is a widely day), they are not willing to sacrifice efficacy for conven-
acceptable method for contaminant detection, due to its ience. Convenience only becomes important when the
high sensitivity and its ability to interface with gas and liquid efficacy and safety characteristics of a drug are similar.
chromatographic techniques (Norwood et al. 2005). Finally, pricing the product the right way by matching it
All parenteral preparations must be free from microbial with the consumer’s buying capability is an important fac-
contamination and their by-products, the endotoxins. tor, in particular when efficacy and safety profiles are
Several tests are available for testing for endotoxins during comparable (Eatmon and Loxley 2009). Another term to
in-process and at the final stage before releasing them to consider beside ESCP is “quality.” This is important when
distributors. Among those tests are the Limulus amebocyte it comes to healthcare issues, where the market demands
lysate test, monocyte activation test, and recombinant fac- quality pharmaceutical/biopharmaceutical products to be
tor C test (Scott and Komski 2020). produced in a most efficient way (Scott 2019). From the
Contamination presents in metered-dose inhalers is FDA’s point of view, when evaluating product quality,
another challenge facing the industry. Although contami- the FDA believes that “quality cannot be tested into the
nation can be traced to ingredients used in the product con- product, but rather it should be built into the product”
centrate, the containers themselves may serve as the main during development (Dziki 2008), often through clear
source. Inhaling contaminants into the lungs allows easy communication between R&D and the product’s manu-
access of potentially toxic materials to the blood circulation facturing unit. For instance, when it comes to the quality
via the alveolar sacs. Thus, proper control of extractables of products intended for pediatric administration, issues
and leachables from containers is an important safety con- such as heavy metals or residual solvents and the presence
cern (Hansen 2005). Most important is screening for sub- of suspected or known genotoxic substances in the
stances such as nitrosamines and polynuclear aromatic formulation must be addressed by the manufacturer
hydrocarbons, as their presence is related to serious adverse (Huang 2020b).
health concerns in humans. Quality control methods are
important for detecting the presence of these substances
at concentrations of nanograms per unit weight of the con- References
tainer material or lower. The leachables profile over the
shelf life of the product should be determined, and agree- Al-Achi, A. and Greenwood, R. (1993a). Human insulin
ment with the level and type of extractables must be estab- binding to erythrocyte-membrane. Drug Dev. Ind. Pharm.
lished (Hansen 2005). 19 (6): 673–684.
Although all impurities in pharmaceutical preparations Al-Achi, A. and Greenwood, R. (1993b). Intraduodenal
are of concern, those found in biologicals are in particular administration of biocarrier-insulin systems. Drug Dev. Ind.
worrisome. Biological impurities result from the presence Pharm. 19 (11): 1303–1315.
of undesirable protein components or nucleic acids that Al-Achi, A. and Greenwood, R. (1993c). Buccal administration
can cause allergic reactions and can lead to mutagenicity of human insulin in streptozocin-diabetic rats. Res.
and carcinogenicity reactions. In particular, these concerns Commun. Chem. Pathol. Pharmacol. 82 (3): 297–306.
are essential to consider if the product is not intended for Al-Achi, A. and Greenwood, R. (1994). Human insulin
single use (Ciurczak 2020). absorption from the intestine in diabetic rats. Drug Dev. Ind.
Pharm. 20 (14): 2333–2339.
Al-Achi, A. and Greenwood, R. (1995). Diffusion profile of
human insulin through polycarbonate membrane. Drug
11.11 Other Considerations Dev. Ind. Pharm. 21 (8): 973–981.
Al-Achi, A. and Greenwood, R. (1998). Erythrocytes as oral
Certain parameters define the “value” of a product. These delivery systems for human insulin. Drug Dev. Ind. Pharm.
are included in the acronym ESCP: efficacy, safety, conven- 24 (1): 67–72.
ience, and price (Eatmon and Loxley 2009). It is the efficacy Al-Achi, A. and Parekh, B. (2004). Preparation and
of a drug that makes it useful clinically. Among drugs with characterization of a compounded aqueous human insulin
similar efficacy, the one with the safest profile receives suspension. Int. J. Pharm. Compd. 8 (6): 492–495.
favorable acceptance among patients and clinicians alike. Al-Achi, A., Greenwood, R., and Harris, T. (1998). Influence of
If efficacy is more crucial than safety (such as the case with pH on the particle size of human insulin. Pharm. Eng. 18 (4):
chemotherapeutic agents), clinicians are willing to use 28, 30, 32.
drugs that are more efficacious, despite pronounced toxic Al-Achi, A., Clark, T.J. III, Greenwood, R., and Sipho Mafu, S.
effect. Although patients prefer the use of products that (2003). Human insulin interaction with soybean powder.
are more convenient to take (e.g. once vs. three times a Pharm. Eng. 23 (1): 40–45.
180 11 Formulation Development Concepts

Al-Achi, A., Patel, J., and Anumandla, M. (2006). Human Bilati, U., Allémann, E., and Doelker, E. (2005). Protein drugs
insulin stability with proteolytic enzymes: the effect of entrapped within micro- and nanoparticles: an overview of
aqueous soybean extract in the formulation. Drug Deliv. therapeutic challenges and scientific issues. Drug Deliv.
Technol. 6 (5): 60–64. Technol. 5 (8): 40, 42–47.
Al-Achi, A., Nguyen, I., Jadhav, R. et al. (2011). Human insulin Brittain, G.H. (1999). What is pharmaceutical physics? Pharm.
diffusion profile from a gel formulation through hair-less Technol. 23 (9): 132, 134, 136, 138.
mouse skin: influence of permeation enhancers. Int. J. Drug Brooks, K. (2020). Keeping up with continuous pharmaceutical
Discov. Herbal Res. 1 (3): 106–112. processing. Contract Pharma ONLINE EXCLUSIVES
Al-Achi, A., Patel, B., and Patel, S. (2012). Oral administration https://www.contractpharma.com/contents/view_online-
of an insulin-soybean suspension in streptozocin rats: effect exclusives/2020-09-11/keeping-up-with-continuous-
of aqueous soybean extract vehicle. Drug Dev. Deliv. 12 (1): processing?utm_source=SilverpopMailing
32–37. &utm_medium=email&utm_campaign=CP_Week+in
Al-Achi, A., Xia, T., and Chaudhari, P.M. (2019). Investigation +Review+%289-11-2020%29+%28Final%29&utm_content=
on complexation efficiency of HP-β-Cd/ nifedipine complex: &spMailingID= 6011722&spUserID =ODg0MzQyMjA5N
pH effect. J. Pharm. Pract. Pharm. Sci. 1: 1–12. TMS1&spJobID =1100512992&spReportId=MTEwMDU
Ali, S. and Quadir, A. (2007). High molecular weight povidone xMjk5MgS2 (accessed 9 November 2020).
polymer-based films for fast- dissolving drug delivery Brower, K.P., Patil, R.A., Walther, J.L. et al. (2020). A tool for
applications. Drug Deliv. Technol. 7 (6): 36, 38–40, 42–43. effective and transport decision-making and efficient
Álvarez, G.D., Esteban, E., Abmus, M., and Skalsky, B. (2007). process development. BioProcess Int. 18 (1-2): 31-
Comparison of Eudragit® FS 30 D and Eudragit® L 30 D-55 34,36-37,47.
as matrix formers in sustained-release tablets. Drug Deliv. Brown, A.-S.B. and Hamed, E. (2007). OraVescent® technology
Technol. 7 (7): 30–35. offers greater oral transmucosal delivery. Drug Deliv.
Amin, A., Shah, T., Parikh, D., and Shah, M. (2008a). Progress Technol. 7 (7): 42–45.
in swellable and low-density excipients for prolonged gastric Brown, M., Lenn, J., and Drummond, J. (2019). Topical
retention. Drug Deliv. Technol. 8 (2): 22, 24–26. development: fast tracking your way to success. Drug Dev.
Amin, A., Shah, T., Patel, J., and Gajjar, A. (2008b). Annual Deliv. 1 (4): 65–69.
update on non-invasive insulin delivery technologies. Drug Browne, J. and Savla, R. (2019). Softgel technology for fast-
Deliv. Technol. 8 (3): 43–48. tracked development programs. Drug Dev. Deliv. 19 (4):
Andrews, R.R. and McClain, J.B. (2008). Breaking through the 31–35.
stent-coating process. Drug Deliv. Technol. 8 (2): 28–31. Calle, S. (2020). Advancements in the field of vaccines and
Anonymous (2018). Hybrid medicines and 505(b)(2) NDA immunization. J. Vaccines Vaccin. 11: e418. https://doi.org/
approval pathways. Altascientist (6): https://www. 10.35248/2157-7560.20.11.e418. © 2020 Calle S. This is an
altasciences.com/sites/default/files/2019-02/The- open-access article distributed under the terms of the
Altascientist_505b2.pdf. Creative Commons Attribution License, which permits
Anonymous (2020a). Fluid handling considerations. BioPharm unrestricted use, distribution, and reproduction in any
Int. 33 (5): 28–31. medium, provided the original author and source are
Anonymous (2020b). The brave new world of manufacturing: credited. (Adopted with modifications.).
speed, capacity, and quality. Gen. Eng. Biotechnol. News Castillo, J. (2013). The vaccine scene. BioPharm Int. 26 (2):
40 (6): 35. 18,20,57.
Arrivo, S.M. (2003). The role of PAT in pharmaceutical Castner, J.C., Williams, N., and Bresnick, M. (2004).
research and development. Am. Pharm. Rev. 6 (2): 46, Leachables found in parenteral drug products. Am. Pharm.
48–49, 51–53. Rev. 7 (2): 70, 72–75.
Aziz, K.J. (2020). Biosimilar biological products: Catalent Zydis brochure (2007). http://www.catalent.com/
development & applications. Drug Dev. Deliv. 20 (2): 24–29. drug/oral/zydis/zydis.pdf; 1–6 (accessed 19 February 2012).
Baluja, N. (2018). Exploring the potential of combining cancer Challener, C.A. (2020). Strategizing for rapid changeovers
vaccines with immune checkpoint inhibitor therapy. Drug in biologics manufacturing. BioPharm Int. 33 (9):
Dev. Deliv. 18 (8): 21–23. 22-25,50.
Bassart, J. (2020). Drug development times, what it takes – Part Chan, P.C. (2008). Biochemical and biophysical methods
1. Drug Dev. Deliv. 20 (1): 28–30. currently used for therapeutic protein development. Am.
Behlke, M. (2020). The importance of assessing off-target Pharm. Rev. 11 (7): 48, 50–54.
effects of CRISPR gene editing. Drug Dev. Deliv. 20 (6): 2629. Chattopadhyay, P. and Shekunov, Y.B. (2006). New enabling
Bellare, J. and Daga, V.K. (2005). Liposome-mediated gene technologies for drug delivery. Drug Deliv. Technol. 6 (8):
delivery. Drug Deliv. Technol. 5 (8): 60–67. 64–68.
References 181

Chaubal, V.M. (2005). Human serum albumin as a Dubin, C.H. (2019). Excipients: formulators want excipients for
pharmaceutical excipient. Drug Deliv. Technol. solubility& beyond. Drug Deliv. Technol. 19 (3): 47–51.
5 (8): 22–23. Dziki, W. (2008). Process analytical technology in product
Cheng, W. and Lim, L.-Y. (2008). Peroral delivery of peptide development and its impact on pre-approval inspections.
drugs. Am. Pharm. Rev. 11 (4): 33–34, 36–41. Am. Pharm. Rev. 11 (7): 12, 14, 16, 18, 20–22.
Chien, W.Y. (1985). The use of biocompatible polymers in rate- Eatmon, C. and Loxley, A. (2009). Chitosan sources, chemistry,
controlled drug delivery systems. Pharm. Technol. 9 (5): 50, properties and pharmaceutical uses. Drug Deliv. Technol.
52, 54, 56, 58, 60, 62, 64, 66. 9 (1): 18, 20, 22–23.
Chowhan, T.Z. (1988). Aspects of granulation scale-up in high- Emisphere Technologies (2012). http://www.emisphere.com/
shear mixers. Pharm. Technol. 12 (2): 26, 28–29, 32, 34, 40, contact.html (accessed 19 February 2012).
42, 44. Endres, P. and Dabre, R. (2020). Reduce downstream
Chowhan, Z.T. and Chi, L.-H. (1985a). Drug–excipient processing costs for mAbs by switching to two-step platform.
interactions resulting from powder mixing: I. Possible BioProcess Int. 18 (6): 66–67.
mechanism of interaction with starch and its effect on drug Erickson, M. (2005). Identification and control of impurities in
dissolution. Pharm. Technol. 9 (3): 84, 86, 90, 92, 94–97. excipients. Am. Pharm. Rev. 8 (1): 88, 90, 92–94.
Chowhan, Z.T. and Chi, L.-H. (1985b). Drug–excipient Eurand N.V. (2012). http://www.aptalispharma.com/en/
interactions resulting from powder mixing: II. Possible pharma_technologies (accessed 21 February 2012).
mechanism of interaction with crospovidone and its effect Evonik Röhm GmbH, Darmstadt, Germany (2012). http://
on in vitro dissolution. Pharm. Technol. 9 (4): 28, 30, 32–33, www.pharma-polymers.com/NR/rdonlyres/3D19A053-
36, 38–41. 7628-41B1-BC52-123763B75566/0/080620FinalProdukt
Chrai, S.S., Murari, R., and Ahmad, I. (2002). Liposomes: a brosch%C3%BCre.pdf (accessed 19 February 2012).
review – I: manufacturing issues. Pharm. Technol. 16: 28, 30, Fabian, L., Hamill, N., Eccles, K.S. et al. (2011). Cocrystals of
32, 34. fenamic acids with nicotinamide. Cryst. Growth Des. 11 (8):
CIMA Labs, Inc (2012). http://www.cimalabs.com/ 3522–3528.
technology/orasolv (accessed 19 February 2012). Fan, Y.F., Wang, Y.N., Fan, Y.G., and Ma, J.B. (2006).
Ciurczak, E.W. (2020). Evaluating biosimilars: a view from the Preparation of insulin nanoparticles and their encapsulation
small-molecule world. BioProcess Int. 18 (1-2): 16–19. with biodegradable polyelectrolytes via the layer-by-layer
Conley, R., Gupta, K.S., and Sathyan, G. (2006). Clinical adsorption. Int. J. Pharm. 324 (2): 158–167.
spectrum of the osmotic-controlled release oral delivery Felgner, P.L., Gadek, T.R., Holm, M. et al. (1987). Lipofection:
system (OROS), an advanced oral delivery form. Curr. Med. a highly efficient, lipid mediated DNA-transfection
Res. Opin. 22 (10): 1879–1892. procedure. Proc. Natl. Acad. Sci. 84: 7413–7417.
Das, T. and Nema, S. (2008). Protein particulate issues in Ford, C.M. (2008). Investment trends driving combination
biologics development. Am. Pharm. Rev. 11 (4): 52, 54–57. product development. Drug Deliv. Technol. 8 (4): 36, 38–39.
Davidson, R. and Rousset, J. (2018). Buccal films: better drug Franzé, S., Selmin, F., Samaritani, E. et al. (2018).
release & patient experiences with buccal films. Drug Dev. Lyophilization of liposomal formulations: still necessary,
Deliv. 18 (7): 22–25. still challenging. Pharmaceutics 10: 139. © 2018 by the
Dean, A.D. (1985). Sources of particulate contamination in authors. Licensee MDPI, Basel, Switzerland. This article is
sterile packaging. Pharm. Technol. 9 (8): 32–36. an open access article distributed under the terms and
Douthwaite, A. (2020). Why pharmaceutical companies are conditions of the Creative Commons Attribution (CC BY)
vulnerable to cyberattacks & what you can do to protect your license (http://creativecommons.org/licenses/by/4.0/).
company. Drug Dev. Deliv. 20 (1): 56–59. (Adapted with modifications.).
Dubin, C.H. (2005a). Formulation frustrations. Drug Deliv. Genzyme Pharmaceuticals (2012). http://www.
Technol. 5 (8): 30, 32, 34. genzymepharmaceuticals.com (accessed 21 February 2012).
Dubin, C.H. (2005b). Combination products: strategies for a Gore, Y.A., McFarland, W.D., and Batuyios, H.N. (1985). Fluid-
continuous pipeline. Drug Deliv. Technol. 5 (9): 40, 42, 44, 46. bed granulation: factors affecting the process in laboratory
Dubin, C.H. (2006). Should the molecule dictate drug delivery? development and production scale-up. Pharm. Technol.
Drug Deliv. Technol. 6 (1): 26, 28, 30–31. 9 (9): 114–116, 118, 120, 122.
Dubin, C.H. (2007). Discussing gastroretentive drug delivery Gothoskar, A.V. (2005). Study of softening point of swellable
systems. Drug Deliv. Technol. 7 (6): 26, 28–29. matrices and effect on drug-release pattern. Drug Deliv.
Dubin, C.H. (2008). Formulating proteins and peptides: Technol. 5 (2): 56–62.
designing for delivery. Drug Deliv. Technol. 8 (2): 34, 36–38. Greenwood, R. and Al-Achi, A. (1997). Human insulin
Dubin, C.H. (2009). On the rise: drug delivery companies you diffusion profile through a layer of Caco-2 cells. Drug Dev.
should know about. Drug Deliv. Technol. 9 (1): 36, 38–43. Ind. Pharm. 23 (2): 221–224.
182 11 Formulation Development Concepts

Guimarães, D., Noro, J., Silva, C. et al. (2019). Protective effect J. Chem. 2019: 9502856, 14 pages. https://doi.org/10.1155/
of saccharides on freeze-dried liposomes encapsulating 2019/9502856. Copyright © 2019 Hidenori Kawasaki et al. is
drugs. Bioeng. Biotechnol. 7: 424. an open access article distributed under the Creative
Gupta, V., Verma, S., Nanda, A., and Nanda, S. (2005). Commons Attribution License, which permits unrestricted
Osmotically controlled drug delivery. Drug Deliv. Technol. use, distribution, and reproduction in any medium,
5 (7): 68–76. provided the original work is properly cited. (Adapted with
Hamilton, L.E. and Lutz, M.E. (2005). Advanced orally modifications.).
disintegrating tablets bring significant benefits to Krishnamurthy, R. (1999). Protein stability in pulmonary
patients and product life cycles. Drug Deliv. Technol. 5 (1): delivery formulations: a review. Pharm. Technol. 23 (3): 48,
34, 36–37. 50, 52, 54, 56, 58, 60.
Hansen, J.K. (2005). Developing a correlation between Langer, R. (1985). Biomaterials: new perspectives on their use
extractables and leachables in MDIs and DPIs. Drug Deliv. in the controlled delivery of polypeptides. Pharm. Technol.
Technol. 5 (2): 66–68. 9 (10): 35, 38, 40.
Hariharan, M. and Bogue, B.A. (2009). Orally dissolving film Langoth, N. and Bernkop-Schnürch, A. (2005). The use of
strips (ODFS): the final evolution of orally dissolving dosage multifunctional polymers as auxiliary agents in non-
forms. Drug Deliv. Technol. 9 (2): 24–29. invasive peptide delivery. Am. Pharm. Rev. 8 (2): 80–84.
Hilleke, K. and Kars, D. (2019). Monetizing innovations in the Laskina, O. (2020). Mitigating risk in pharmaceutical
biopharmaceutical industry. BioProcess Int. 17 (1-2): manufacturing with visually inspected components. Drug
14,16-17. Dev. Deliv. 20 (2): 52–55.
Hincha, D.K., Hellwege, E.M., Heyer, A.G., and Crowe, J.H. Le Gal, P. (2019). Quality-by-Design approach to enable high-
(2000). Plant fructans stabilize phosphatidylcholine dose drug delivery with autoinjectors. Drug Dev. Deliv. 19 (5):
liposomes during freeze-drying. Eur. J. Biochem. 34–39.
267: 535–540. Leveen, L. (2010). Pre-sterilized, single use filling systems for
Holm, R. (2010). Enabling pharmaceutical technology lipid liquid bio-pharmaceuticals. Am. Pharm. Rev. 13 (5): 30,
based formulations. Am. Pharm. Rev. 13 (5): 10, 12–15. 32, 33.
Huang, J. (2019). Revitalization of older drug products using Lingelbach, D. (2020). The role of viral vectors play in current
innovation formulation technologies by 505(b)(2) regulatory gene therapy development. Drug Dev. Deliv. 20 (5):
pathway. Drug Dev. Technol. 19 (3): 16–18. 50–54.
Huang, J. (2020a). Application of nano-emulsion technology to Lipocine, Inc (2012). http://www.lipocine.com/nanosplode.
address unmet medical needs: a case study of clopidogrel IV html (accessed 19 February 2012).
by 505(b)(2) pathway. Drug Dev. Technol. 20 (4): 30–33. Lopes, C.M. and Soares, C. (2015). Transdermal drug delivery
Huang, J. (2020b). Age-appropriate pediatric formulation systems activated by physical stimuli: Techniques and
development. Drug Dev. Deliv. 20 (8): 18–20. applications. Drug Des. 4: e129. https://doi.org/10.4172/
Jain, K.S., Jain, N.K., and Agrawal, G.P. (2005). Gastroretentive 2169-0138.1000e129. Copyright: © 2015 Lopes CM, et al.
floating drug delivery: an overview. Drug Deliv. Technol. This is an open-access article distributed under the terms of
5 (7): 52, 54–61. the Creative Commons Attribution License, which permits
Jinek, M., Chylinski, K., Fonfara, I. et al. (2012). A unrestricted use, distribution, and reproduction in any
programmable dual-RNA-guided DNA endonuclease in medium, provided the original author and source are
adaptive bacterial immunity. Science 337: 816–821. credited. (Adapted with modifications.).
Joshi, A.A. and Duriez, X. (2004). Added functionality Lowe, D., Mehta, M., Govindan, G., and Gupta, K. (2018).
excipients: an answer to challenging formulations. Pharm. Enabling flexibility of supply chain and drug-product
Technol. (Excipients and Solid Dosage Forms) 12–19. presentation for biologics. BioProcess Int. (Featured Report)
Kannan, V., Balabathula, P., Thoma, L.A., and Wood, G.C. 16 (11-12/A Special Insert): 8–13.
(2015). Effect of sucrose as a lyoprotectant on the integrity of Lynch, P.M. (2011). Extractables and leachables: quality
paclitaxel-loaded liposomes during lyophilization. J. concerns and considerations for ophthalmic and injectable
Liposome Res. 25 (4): 270–278. https://doi.org/10.3109/ products. Am. Pharm. Rev. 14 (4): 93–94, 96.
08982104.2014.992023. Majithiya, R.J., Raval, A.J., Umrethia, M.L. et al. (2008).
Kantor, A., Tchessalov, S., and Warne, N. (2011). Quality-by- Enhancement of mucoadhesion by blending anionic,
design for freeze–thaw of biologics: concept and application cationic and nonionic polymers. Drug Deliv. Technol. 8 (2):
to bottles of drug substance. Am. Pharm. Rev. 14 (4): 40–45.
65–66, 68–72. Majuru, S. (2005). Development of an oral insulin solid dosage
Kawasaki, H., Shimanouchi, T., and Kimura, Y. (2019). Recent formulation using emisphere’s eligen® technology. Drug
development of optimization of lyophilization process. Deliv. Technol. 5 (9): 74, 76–78.
References 183

Manley, L., Hilden, J., Valero, P., and Kramer, T. (2019). Tablet formulation of remdesivir in treating COVID-19. Drug Dev.
compression force as a process analytical technology (PAT): Deliv. 20 (5): 1–8.
100% inspection and control of tablet weight uniformity. Plotkin, S., Robinson, J.M., Cunningham, G. et al. (2017).
J. Pharm. Sci. 108: 485–493. The complexity and cost of vaccine manufacturing – an
Markarian, J. (2020). Preparing pandemic vaccine capacity. overview. Vaccine 35 (33): 4064–4071.
BioPharm Int. 33 (9): 10–14. Pondell, R. (1985). Scale-up of film coating processes. Pharm.
Mehta, M.A. (1988). Scale-up considerations in the fluid-bed Technol. 9 (6): 68, 70.
process for controlled-release products. Pharm. Technol. Porter, C.S. and Saraceni, K. (1988). Opportunities for cost
12 (2): 46–47, 50–52. containment in aqueous film coating. Pharm. Technol.
Menezes, J.C., Testas, M.R., Santos, S.T. et al. (2019). Is the 12 (9): 62, 64, 66, 68, 70, 74, 76.
QbD toolbox ready for cell and gene therapies? Integrating Prajapati, B.G., Patel, J.K., Patel, V.M., and Prajapati, K.V.
patient outcomes into manufacturing cell and gene therapy (2008). The upcoming era of nanomedicine: a briefing. Drug
bioproducts. BioProcess Int. 17 (4): 56–58. Deliv. Technol. 8 (4): 46, 48–49.
Merrion Pharmaceuticals (2012). http://www. Rajan, M.G., Bhattacharjee, H., Bedi, S., and Reo, J. (2010).
merrionpharma.com/content/technology/gipet.asp Primer on NIR spectroscopy as a PAT tool in the tablet
(accessed 19 February 2012). manufacture process. Am. Pharm. Rev. 13: 128–135.
Mirasol, F. (2020). Single-use bioreactors: to scale up or scale Ravishankar, H., Patil, P., Petereit, H.-U., and Renner, G.
out? BioPharm Int. 33 (1): 19–21. (2005). EudramodeTM: a novel approach to sustained oral
Myatt, R., Oladiran, G., and Batchelor, H. (2008). Measuring drug delivery systems. Drug Deliv. Technol. 5 (9):
the solubility of a model drug in drug-in-adhesive 48, 50, 52–55.
transdermal patches to validate a theoretical solubility Robinson, J.M. (2009). An alternative to the scale-up and
calculator. Drug Deliv. Technol. 8 (3): 56, 58–59. distribution of pandemic influenza vaccine. BioPharm Int.
Navarro, F. (2019). Lipid-based system introduces a novel 2009 (Supplement, Issue 1): 40–45.
approach for an HIV vaccination. Drug Dev. Deliv. 19 (6): Russo, J.E. (1984). Typical scale-up problems and experiences.
16,18,20-21. Pharm. Technol. 8 (11): 46–48, 50–51, 54–56.
Northup, S.J. (2008). Pharmaceutical containers: safety Sacha, G.A. (2020). Process monitoring during freeze-drying.
qualification of extractables/leachables. Am. Pharm. Rev. Drug Dev. Deliv. 20 (4): 34–39.
11 (3): 104, 106, 108–109. Saharan, P. (2019). Exhibition on pharmaceutics & novel drug
Norwood, D.L. and Mullis, J.O. (2009). “Special case” delivery systems- market analysis. Am. J. Adv. Drug Deliv.
leachables: a brief review. Am. Pharm. Rev. 12 (3): 78, 7 (1): 1–3. (21st International Conference and Exhibition on
80, 82–86. Pharmaceutics & Novel Drug Delivery Systems - March
Norwood, D.L., Nagao, L., Lyapustina, S., and Munos, M. 11-12, 2020 Rome, Italy; Future Outlook and New Era
(2005). Application of modern analytical technologies to the Technologies in Pharmaceutics and Novel Drug delivery
identification of extractables and leachables. Am. Pharm. Systems).
Rev. 8 (1): 78, 80, 82–87. Sarah Motter (2020). “13WIBW” TV channel, Published:
O’Connor, D.J. (2019). Arming the immune system; turning 8 June 2020, at 12:37 p.m. EDT. https://www.wibw.com/
cold tumors hot. Drug Dev. Deliv. 19 (6): 42,43-46. content/news/KU-has-heavy-hand-in-creation-of-
Okutgen, E., Orhan, A., and Bassart, J. (2020). PEGylation – Remdesivir-571103991.html (accessed 18 December 2020).
three decades of product approvals & technology Sawant, P.D., Luu, D., Ye, R., and Buchta, R. (2010). Drug
development. Drug Dev. Deliv. 20 (7): 24–27. release from hydroethanolic gels: effect of drug’s
Olsen, W.K. (1989). Batch fluid-bed processing equipment – a lipophilicity (logP), polymer–drug interactions and solvent
design overview: I. Pharm. Technol. 13 (1): 34, 36, 38, 40, 42, lipophilicity. Int. J. Pharm. 396 (1–2): 45–52.
44, 46. Scarlett, B. (2003). Measuring and interpreting particle size
Panjwani, L. (2019). Fighting the flu: the quest for a better distribution. Am. Pharm. Rev. 6 (4): 93–101.
vaccine. R&D Magazine 61 (2): 12–16. Scott, C. (2019). Biomanufacturers need smart strategies to
Patel, V.F. and Patel, N.M. (2008). Influence of drug-to-lipid speed products to market. BioProcess Int. 17 (1-2/Featured
ratio and release modifier on dipyridamole release from Report): 6–12.
floating lipid granules. Drug Deliv. Technol. 8 (4): 22, Scott, C. and Komski, L. (2020). Endotoxin questions and
24, 26–27. answers. BioProcess Int. 18 (11-12): 56–57.
Patel, G., Patel, G., Patel, R. et al. (2007). Sodium alginate: Scott, C., Hauk, A., and Tappe, A. (2020). The proof is in the
physiological activity, usage and potential applications. data: extractables and leachables. BioProcess Int. 18 (1-2):
Drug Deliv. Technol. 7 (4): 28, 30, 32, 34, 36–37. 42–44.
Pipkin, J., Antle, V., and García-Fandiño, R. (2020). Shaffer, C. (2020). Gene therapy startups: extended biotech’s
Application of Captisol® technology to enable the legacy. Gen. Eng.Biotechnol. News 40 (10): 32,34,36.
184 11 Formulation Development Concepts

Shankar, B. (2005). Insulin delivery technologies: present and Timpe, C. (2007). Strategic for formulation development of
future. Drug Deliv. Technol. 5 (9): 38–39. poorly water-soluble drug candidate: a recent perspective.
Sirisha, V. and Sailaja, A.K. (2018). Review on recent Am. Pharm. Rev. 10 (3): 104, 106–109.
approaches in transdermal drug delivery system. J. Nurs. Tingstad, E.J. (1988). The key to effective communication.
Patient Health Care 1 (1): 103. Copyright: © 2018 Sirisha V. Pharm. Technol. 12 (10): 30, 32.
This is an open-access article distributed under the terms of Vela Ramirez, J.E., Sharpe, L.A., and Peppas, N.A. (2017).
the Creative Commons Attribution License, which permits Current state and challenges in developing oral vaccines.
unrestricted use, distribution, and reproduction in any Adv. Drug Deliv. Rev. 114: 116–131.
medium, provided the original author and source are Washer, S. and Knop, D.R. (2020). Viral-vectored gene therapies:
credited. (Adapted with modifications.). harnessing their potential through scalable, reproducible
Smith, M. (2020). A new era: why the time has come for manufacturing processes. BioProcess Int. 18 (6): 46,48,50.
biopharma too move to continuous bioprocessing. Drug Dev. Wei Ling, H. (2020). World vaccines and immunology market
Deliv. 20 (1): 42–45. analysis. J. Vaccines Vaccin. 11 (1) This is an open access
Stagner, W.C., Jain, A., Al-Achi, A., and Haware, R.V. (2020). article distributed under the terms of the Creative Commons
Employing multivariate statistics and latent variable models Attribution License, which permits unrestricted use,
to identify and quantify complex relationships in typical distribution, and reproduction in any medium, provided the
compression studies. AAPS PharmSciTech 21: 186. https:// original work is properly cited.: https://www.
doi.org/10.1208/s12249-020-01712-1. walshmedicalmedia.com/open-access/world-vaccines-and-
Stegemann S. (2020). Beyond the boundaries: new capsule immunology-market-analysis.pdf.
types resolve emerging formulation needs. Contract Wen, Z.-Q., Cao, X., and Philips, J. (2010). Application of
Pharma. https://www.contractpharma.com/issues/2020-09- Raman spectroscopy in biopharmaceutical manufacturing.
01/view_features/beyond-the-boundaries/ (accessed 9 Am. Pharm. Rev. 13 (4): 46, 48, 50–53.
November 2020). Wiggins, J.M. and Albanese, J.A. (2020). Pharmacopoeia
Straiton, J. (2020). CRISPR vs COVID-19: how can gene editing compliance: a practical guide. BioPharm Int. 33 (9): 40–45.
help beat a virus? Biotechniques 69 (4-6): 75–77. Wolf, J. (2020). COVID-19 vaccine focusing on T cells to protect
Tchessalov, S., Dixon, D., and Warne, N. (2007). Principles of the most vulnerable. Drug Dev. Deliv. 20 (8): 31–34.
lyophilization cycle scale-up. Am. Pharm. Rev. 10 (3): 88–92. Wormuth, K., Gauthier, M., Labedan, M. et al. (2019). Visible
Thapa, R.K., Winther-Larsen, H.C., Diep, D.B., and Tønnesen, particulate matter in single-use bags. BioProcess Int. 17 (4):
H.H. (2020). Preformulation studies on novel garvicin KS 50–53.
peptides for topical applications. Eur. J. Pharm. Sci. 151: Zelesky, T. (2008). Supercritical fluid chromatography (SFC) as
105333. © 2020 The Authors. Published by Elsevier B.V. This an isolation tool for the identification of drug related
is an open access article under the CC BY license (http:// impurities. Am. Pharm. Rev. 11 (7): 56–60, 62.
creativecommons.org/licenses/BY/4.0/). (Adapted with Zhang, S., Goldman, J., Chen, X. et al. (2020). Non-invasive,
modification.). quantitative characterization of lyophilized drug product
Thomas, F. (2020a). Advancing understanding of a traditional using three-dimensional x-ray microscopy analytics. Drug
technique. PharmTech 44 (9): 44–49. Dev. Deliv. 20 (1): 32–40.
Thomas, S. (2020b). Capsule innovations: speeding up drug
development. PharmTech 44 (9): 30–32.

Glossary
API Active pharmaceutical GRAS Generally recognized as safe.
ingredient.
HSA Human serum albumin.
BBB Blood–brain barrier.
HLB Hydrophile–lipophile balance.
CNS Central nervous system.
HPLC High-performance liquid
FDA US Food and Drug chromatography.
Administration.
HPMC Hydroxypropyl methylcellulose.
GI Gastrointestinal.
Industrial Both preformulation and formula-
GIPET Gastrointestinal permeation pharmacy tion are parts of pharmaceutics,
enhancement technology. and their application in

You might also like