You are on page 1of 13

Journal of Pharmaceutical Sciences 109 (2020) 3235-3247

Contents lists available at ScienceDirect

Journal of Pharmaceutical Sciences


journal homepage: www.jpharmsci.org

Review

Freeze-Drying of Pharmaceutical and Nutraceutical Nanoparticles:


The Effects of Formulation and Technique Parameters on
Nanoparticles Characteristics
Mohsen Mohammady a, Yasaman mohammadi a, Gholamhossein Yousefi a, b, *
a
Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, P.O. Box 71345-1583, Shiraz, Iran
b
Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran

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

Article history: Nanoparticles (NPs) are of the most interesting novel vehicles for effective drug delivery to humans.
Received 6 April 2020 Freeze drying is known as an engaging process to improve the long lasting stability of NPs formulations.
Revised 23 May 2020 This study aims to elucidate the importance of various parameters involving in freeze-drying of the most
Accepted 15 July 2020
common pharmaceutical/nutraceutical NPs including nanosuspensions, nanocrystals (NCs), cocrystals/
Available online 20 July 2020
nanococrystals, nanoemulsions (NEs), nanocapsules (NCPs) and nanospheres (NSPs). Regarding this, the
therapeutic goals of NPs and specifications of drug must be considered. According to our survey, the most
Keywords:
Nanoparticles influential factors for achieving optimum results include type and concentration of cryoprotectant/lyo-
Lyophilization protectant, stabilizer structure and concentration, the NPs concentration in solution, freezing, annealing,
Nanocrystal and drying rate, the interaction between protectants and stabilizer, solvent type and antisolvent to
Nanococrystals
solvent ratio. The study shows that for each class of NPs, specific variables are of highest significance and
Nanoemulsion
Nanospheres should be optimized. For instance, about NCs, freezing rate and antisolvent/solvent ratio should be
Nanosuspension particularly considered and for emulsified NPs, the best results have been obtained by 5e20% of sac-
charides as cryoprotectants. These findings suggest that to obtain a product with the lowest aggregation
and particle size (PS), optimization of the effective factors in formulation and lyophilization process are
essential.
®
© 2020 American Pharmacists Association . Published by Elsevier Inc. All rights reserved.

Introduction improve drug permeation through biological barriers for instance,


intestinal and respiratory epithelia.5,6 NPs are mostly fabricated in
Nanoparticles aqueous media. However, these aqueous dispersions are not actu-
ally stable making them susceptible to aggregation and fusion of
The drug delivery to humans based on NPs is of the most particles throughout long periods of storage.7 Additionally, the
interesting strategies recently employed for achieving more effec- other physical stability difficulties are common with NPs including
tive drug therapy with lower side effects in various administration sedimentation, creaming (the tendency of oil droplets for aggre-
routes.1 Generally, the word “NP” refers to particles with sizes in the gating to diminish the surface energy), agglomeration, crystal
range of 1e100 nm. However, in the field of pharmaceuticals and growth (Ostwald ripening) and change of crystallinity state
medicine, submicron particles are normally called NPs.2,3 The NPs (changing from amorphous to crystalline).8 The chemical instability
give numerous profits over microparticles. The submicron size of of NPs structure includes the hydrolysis of polymeric materials
NPs causes to higher intracellular uptake compared to micropar- which depends on the conditions in which the NPs are stored such
ticles.4 Routinely, due to their capability to keep the loaded drug as temperature, medium pH and polymer characteristics (for
stable and provide it in a consistent manner, NPs can significantly instance its structure and molecular weight). This kind of instability
enhance the bioavailability of many drugs. Moreover, NPs can is frequently reported during NPs storage for long time.9,10 There-
fore, when developing the NPs formulations, how physically and
chemically stable they remain in long periods of time can be a
detrimental issue.11
* Corresponding author. The Department of Pharmaceutics, School of Pharmacy,
Shiraz University of Medical Sciences, Shiraz, Iran.
Freeze drying is a routinely used industrial process to improve
E-mail address: ghyousefi@sums.ac.ir (G. Yousefi). the long-term chemical stability of pharmaceutical formulations

https://doi.org/10.1016/j.xphs.2020.07.015
0022-3549/© 2020 American Pharmacists Association®. Published by Elsevier Inc. All rights reserved.
3236 M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247

including NPs owing to its unique advantages. Particularly, its to boost stability during lyophilization and also improve physical
possibility of application for small volume products and its rather stability of the product at storage time.23 Excipients used for this
simple scale up are of high significance.12 Freeze-drying is by purpose are divided into two categories of cryoprotectant and
definition a low temperature dehydration process by transition lyoprotectant agents. Cryoprotectants shield the product through
from the solid to the gas state without passing through a liquid the freezing step and lyoprotectants prevent from the stresses
state. It consists of three sequential steps of freezing, primary caused during the drying step. The protecting impact of cryopro-
drying and secondary drying.13 Through the process of sublimation, tectants is described by many elements. As freezing is applied in a
water is eventually eliminated from ice at pressures and tempera- temperature lower than glass transition temperature (Tg) of cryo-
tures below the triple point of water.14 Lyophilized formulations protectant, immobility of particles in its glassy matrix increases the
give straightforward handling during shipping and storage.15 stability of the product.24 Various approaches which protect the
Similar to solid state medicines like pills or capsules, the NPs as NPs native structure, rely on replacing the water. This hypothesis
liquid dispersions should be firstly converted to dry powders and might explain the NPs stabilization by hydrogen bonds between the
next converted to other solid dosage forms if needed.16 In the polar groups at the surface of NPs and stabilizer molecules caused
course of the procedure, freezing and dehydration stresses are by water losing. Therefore, the characteristics of NPs are saved by
developed and they might probably enhance the aggregation of NPs the stabilizers through acting as water replacement. Besides, add-
causing to instability. Different excipients like cryoprotectants and ing cryoprotectant agents that increase the viscosity within the
lyoprotectants could also be utilized to decrease the freeze-drying sample can subdue the construction of ice crystals. Consequently,
stresses, and conserve the physicochemical characteristics of aggregation through the freezing step in temperatures above Tg is
NPs.17 This study aims to obtain an updated insight into the es- averted by the isolation of separate particles.25 The most typical
sentials involving in the lyophilization of pharmaceutical and nu- cryoprotectants are sugars including monosaccharides (glucose,
traceutical NPs including nanosuspensions, NCs, NEs, NCPs and fructose, and galactose), dissaccharides (sucrose, lactose, trehalose,
NSPs. Mainly, it focuses on the formulation considerations and and maltose) and tri-saccharides (Raffinose).
freeze-drying cycle optimizations to obtain a suitable lyophilized Trehalose, due to lack of internal hydrogen bonding, low hy-
product. groscopicity, low chemical reactivity and high Tg, is more efficient
than other sugars.26 For stabilizing the PS of chitosan NPs, glucose,
Freeze Drying Stages sucrose, trehalose, mannitol, polyethylene glycol (PEG) 2000, and
PEG 10000 cryoprotectants at concentrations of 5%, 10%, 20% and
The procedure of freeze drying encompasses three essential 50% were employed. Among these cryoprotectants, the best pro-
steps. First one is freezing or solidification, next is primary drying, tection was observed for sucrose and trehalose at concentration of
also called ice sublimation and finally, secondary drying which is 5% such that there was no growth in the PS due to the lack of ag-
moisture desorption. Primarily in the process of freeze-drying, the gregation. Other cryoprotectants had little effect and there was a
liquid suspension is frozen and consequently, the pure water con- significant rise in the size of the particles. For instance, when PEG
verts into ice crystals. As the process continues, the proportion of 10000 was used as cryoprotectant, particles enlarged significantly
ice crystals and the concentration of the still existing liquid in- in the samples.27 On the other hand, destabilization of NPs by a
crease. Consequently, the viscosity of product increases and this typical cryoprotectant has been reported by some researchers. In
results in inducing restraint for further crystallization. As this vi- the case of increase in the concentration of a cryoprotectant in NPs
cious and concentrated liquid becomes solid, an amorphous, crys- formulation, both raise or reduction in NPs’ aggregation can be
talline, or combined amorphous-crystalline solid will be obtained.18 observed.28
“Bound water” is the small amount of water which does not freeze The protective effect of polyols (glycerol, mannitol, sorbitol,
and remains liquid. Drying step is the process which the ice from adonitol, and inositol) has been in general lower than that observed
the frozen product is sublimated.19 At the first stage, heat is con- for sugars. Mannitol due to its potential for crystallization and also
ducted from the shelf through the tray and subsequently to the forming eutectic mixture with ice will separate the phases within
frozen solution and directed to the subliming front. At the second the cryo-concentrated portion of the frozen NPs without any stable
stage, the sublimed ice changes into vapor and goes through the interactions with them. Separate NPs in the NP-rich phase will
dehydrated part to the top of the sample. At the third stage, the interplay and form aggregates. Furthermore, crystals of water and
water vapor is moved through the chamber to the condenser. At the mannitol which are enlarged may apply mechanical stresses on the
final stage, the water vapor condenses and a permeable plug is NPs leading to their fusion. Therefore, a minimum number of
shaped in which small openings can be seen in the place of ice mannitol molecules should stay molecularly scattered in the
crystals.20 At secondary drying step, the water which is not isolated amorphous NPs phase in a stabilization process.29 X-ray diffraction
as ice nor sublimated off is removed from the product.21 Fig. 1 discovered that monosaccharide-including freeze-dried
depicts the above-mentioned stages of freeze drying process griseofulvin-lipids NPs had crystalline structure. Quite the oppo-
regarding the how of water removal from NPs suspensions. site, disaccharide-including freeze-dried griseofulvin-lipids NPs
showed an amorphous structure.30 Polymers (dextran, starch, PEG,
Cryoprotectants and Lyoprotectants and polyvinyl pyrrolidone) have stabilizing role in NPs. PEGs have
additionally been utilized as stabilizer in freeze drying of poly-
Different stresses are developed throughout freeze-drying pro- electrolyte complex (PEC) NPs as well as chitosan NPs. A rise in
cess. These stresses result into worsening of the texture of the mass trehalose concentration up to 2% w/v caused a reduction in the PS
due to the shock caused by low temperature, osmotic shrinkage and ratio (defined as the ratio of PS after to before freeze-drying) and
particles aggregation when rehydrating.22 Within the cryo- slight PEC NPs aggregation. When PEC NPs were freeze dried with
concentrated phase, high concentration of NPs might induce their 3, 4 or 5% w/v of trehalose, PS ratio decreased and aggregation was
aggregation or fusion. Besides, NPs might lose their stability as a not seen. With 5% w/v trehalose not reduction in PS neither ag-
result of the mechanical pressures produced by ice crystals. To gregation of particles were seen, thus at 5% w/v, trehalose acted in
prevent the mentioned events, several excipients are also included the role of an efficient cryoprotectant for PEC NPs. Also, the use of
into the NPs formulation before freeze-drying.17 In this case, the polymer PEG could be more effective than trehalose so that 1% of
excipients are inert substances added to a pharmaceutical product PEG 2KD in contrast to 3% of trehalose could make redispersibility
M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247 3237

acceptable. Best protection for NPs was obtained by using 2% PEG Nanosuspensions, described as colloidal dispersions of nano-sized
which was comparable to 4% trehalose. The combination of treha- medicine particles, may attain great drug loading efficiency (up to
lose and PEG 2KD as protectant produced a synergistic effect 100%) in drug delivery systems.16 Normally, nanosuspensions made
reducing the PS ratio and the aggregation of NPs. Therefore, it can stable by surfactants or polymeric steric stabilizers and constructed
be concluded that adding a polymer like PEG can reduce the con- by appropriate techniques may cause to a smart stability.34 But, the
centration of the sugar required for lyophilization.31 Therefore, the most of NPs are chemically and physically less stable in aqueous
use of suitable cryoprotectant at optimal concentration results in suspensions. NPs are often formed in an aqueous environment and
significant positive effects in controlling the distribution of PS and this environment causes to physical instability as particle aggre-
shape. The application of trehalose, mannitol, sorbitol or glycerol as gation and fusion over time.7 NP suspensions are also susceptible to
cryoprotectant is a useful method for freeze-drying of NPs to keep the growth of microorganisms.8 To solve this problem, a suspension
up their physical characteristics.32 Consequently, cryo and lyopro- containing a NP should be formulated as a solid. Lyophilization is an
tectant type and concentration, protectant to NPs mass ratio and answer to this problem with the aid of various excipients such as
the possibility of crystal or amorph formation are important vari- cryoprotectants and lyoprotectants which are used to reduce
ables in protectant selection for stabilizing NPs. Fig. 2 demonstrates lyophilization stress and maintain physical and chemical proper-
the role of cryoprotectants in prevention of NPs aggregation after ties.28 As a whole, lyophilized NPs are considerably aggregative and
rehydration of lyophilized NPs. particle dimentions rise once it is reconstituted into solution
despite of having surfactants in their structure.35 The use of sugars
Freeze Drying of Pharmaceutical NPs due to the effect on Tg has an important role in their lyophiliza-
tion.36 It is declared that poly (DL-lactide) -poly(ethylene oxide)
Nanosuspensions (PLA-PEO) NPs have been aggregated during freeze-drying and the
complication as a major drawback could be bypassed by using
The nanosuspensions have extended the innovative and assur- trehalose in the role of a cryoprotectant.37 Optimization of lyoph-
ing capacities for delivery of water-insoluble drugs.33 ilization includes the selection of suitable cryoprotectant, freezing

Fig. 1. The varying stages in freeze drying of nanoparticles and the phase transitions of product water through different stages of the process.
3238 M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247

Fig. 2. The role of protecting agents in prevention of nanoparticles aggregation after reconstitution of lyophilized nanoparticles.

rate, formulation characteristics, and lyophilization cycle.38 the NPs surface.41 In another study, the importance of the combi-
Therefore, decent lyophilization of NPs results in the easy re- nation of the two elements “steric stabilizer concentration” and
suspending of NPs, the minimum change in particle dimensions “cryoprotectant concentration” was disclosed to affect mutually
and distribution, and maintaining the activity of the loaded drug.20 throughout the freezing phase.42,43 Increasing the concentration of
Table 1 summarizes the freeze-dried nanosuspensions found in steric stabilizer lowered the propensity of particles aggregation.
literature including the manufacturing method and main excipi- Raising the steric stabilizer concentration generated a thicker steric
ents used to optimize freeze-drying. layer on the surface of the NPs that separated them within the cryo-
concentrated regions. The obtained results proved that while a rise
Formulation Effect in the steric stabilizer concentration increased the stability of NPs, a
The formulation parameters including polymer structure and rise in the disaccharide concentration along with higher concen-
concentration, type of the surfactant and its concentration, and the trations of steric stabilizer was notably more efficient in reducing
nature and concentration of cryoprotectant and lyoprotectant in the size of particles for nanosuspensions.44
lyophilization are important.38 In order to increase the stability of
NPs and minimize their aggregation, polymers are routinely used. Cryo& Lyo Protectants Effect
Throughout the lyophilization process, these polymers might have Lyophilization may cause mechanical stress on nanosuspensions
an impact on stabilizing NPs. It has been revealed that polyvinyl which to solve this problem, the addition of some excipients as
alcohol (PVA) as a stabilizer can be the reason of small PSs.39 In cryoprotectant and lyoprotectant to the formulation is considered
caprolactone NPs stabilized with 2.5e5% of PVA, no significant as an option. Cryoprotectants prevent aggregation by protecting
aggregation was observed after lyophilization.38 This observation NPs via the matrix mechanism and the hypothesis of separating the
was possibly the consequence of PVA being present on the surface particles in the unfrozen phase.21,25 The most commonly used
of the NPs which affects improvement of freezing.40 However, all of cryoprotectants are sugars with trehalose having more benefits
the stabilizers do not increase the stability of NPs throughout than other sugars. The stability of system depends on the concen-
lyophilization. Additionally, the medicine trapped inside the NPs or tration and mass ratio of cryoprotectant to NPs. It has been argued
free unloaded drug are effective on the lyophilization process. If the that PLA-PEO re-dispersion after lyophilization occurred at a 1:1
drug is free, it reduces zeta potential and leads to aggregation. For mass ratio of trehalose to NPs.37 Another study showed that,
example, lyophilization of cyclosporine NPs in the presence of resveratrol NPs with quercetin as stabilizer and 1% w/w of mannitol
pluronic F68 was improved as a result of free drug adsorption onto and trehalose produced large NPs due to decreased cryoprotectant

Table 1
The Examples of Drug Nanosuspensions Manufacturing Methods and Main Excipients Used for Freeze-Drying.

Compound Method Aid Excipients for Freeze-Drying Literature Reference


28
Naproxen Custom made apparatus (the yttrium-stabilized zirconium beads) Lactose, sucrose, mannitol, PEG 2000
119
Azithromycin HPH Poloxamer 188
120
Carvedilol Anti-solvent precipitatione ultrasonication Maltose
121
Loviride Media milling Sucrose
122
Silymarin Sonoprecipitation Mannitol
123
Danazol Wet milling/HPH Mannitol
124
Flurbiprofen High pressure homogenization e
125
Loratadine Anti-solvent precipitation Sucrose
126
Celecoxib HPH Lactose, sucrose, trehalose or mannitol
M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247 3239

concentrations. However, increasing the concentration of mannitol processes to prepare NCs are often classified as top-down and
and trehalose up to 5% w/w improved re-dispersion of resveratrol bottom-up. High-pressure homogenization53 and also wet ball
NPs.45 Lyoprotectants are required to prevent drying stress by the milling are considered as common top-down processes.54 Draw-
mechanism of forming hydrogen bonds between the lyoprotectants backs of the so called processes can be the utilization of surfactants,
and the polar groups of NPs' surfaces.29 NPs’ concentration in this the lengthy processing times, particle size polydispersity, low
mechanism is important and it has been observed that a high yields, great energy consumption and potential air pollution com-
concentration of NPs resulted in high efficiency of lyoprotectants.46 ing from the means of grinding.55e57 Bottom-up processes are
Besides, it has been argued that re-dispersibility increases by primarily precipitation ones. An important drawback of the most
increasing the molecular weight of cryoprotectant. It was proved commonly used bottom-up processes is the inability to properly
that the aggregation of drug NPs can be avoided more efficiently control the eventual size of drug crystals.57,58 In spite of their
when PEG had a higher molecular weight. On the other hand, PEG benefits, drug NCs have numerous disadvantages such as complex
with the lowest molecular weight (PEG 0.4 k) led to an important scaling up,55,58,59 possibility of nano-toxicities60 and stability
aggregation that was shown as particles which were bigger in size issues.61e63 One of the most crucial points to make sure these drug
relative to initial sizes.13 products are safe and efficient is their chemical stability. Freeze-
drying and spray drying are among the most typical solidification
processes for NCs. Since a great deal of solid NCs are typically
Freeze-Drying Cycle Effect
reconstituted into nanosuspensions, their growing or agglomera-
Freezing is the first step in the process of lyophilization, which is
tion must be avoided through drying process. This consideration
defined by cooling the formulation and forming crystalline ice.
helps to keep up the nano-sized properties of NCs including fast
During freezing, the viscosity of concentrated residual suspension
dissolution after the reconstitution. In order to cope with the sta-
increases and prevents crystallization.47 A solution, when reaches
bility problems during the solidification process, matrix formers
to a temperature of Tg, has the highest concentration of cryo-
like mannitol, sucrose and polysaccharides can be added to NCs
concentrated solution. Above Tg, concentration is not altered by
formulations.62 Liquid NC dispersions designed for administration
cooling, so the freezing temperature should be lower than Tg and be
as solid dosage forms should be freeze-dried to produce nano-sized
adequate for a sufficient period to guarantee the perfect solidifying
crystalline materials.64 An aqueous solution of cryoprotectant is
of suspension.48 There is a controversy about the optimal slow or
mixed with organic drug solution and after nano-precipitation of
fast freezing rate of NPs. Some studies have reported that fast
crystals and organic solvent evaporation, the lyophilization take
freezing has reduced PS compared to slow freezing.49 When pro-
place. The medicine chosen to be used in the technique ought to
tectant is added to the formulation, usually the freezing rate does
have a low degree of glass transition (Tg). In order to reach to the
not affect the PS.50 It has been reported that fast freezing in the
drug crystals, the freeze-drying temperature must be more than
presence of mannitol caused to a smaller PS than slow freezing.30 In
drug's Tg.65 Robust agglomerates can be formed from NCs in the
another study, slow freezing was more efficient for lyophilization
case of unsuitable drying making their re-dispersion difficult. Thus,
regarding size. For example, in the evaluation of the effect of
it is of vital importance that the NPs remain discrete (re-dispers-
cryoprotectant on freezing rate of hydroxypropyl cellulose (HPC),
ible) throughout the drying step.66 Table 2 summarizes the freeze-
slow freezing was proved to be better.28 It was observed that with
dried NCs found in literature including the manufacturing method
varying the rate of freezing and the concentration of cryoprotectant
and main excipients used to optimize freeze-drying.
simultaneously, a faster rate of freezing and a higher concentration
were superior. However, when the freezing rate was relatively slow,
it caused to greatly decrease in the rate of NPs aggregation. The
Lyophilization Process Effect
slower rate of freezing may stimulate the migration of cryopro-
Polymers' steric stability becomes inactive during freeze-drying
tectants to a cryo-concentrated liquid phase, leading to an increase
of NCs dispersions. Through the extension of the freezing interface,
in its native concentration which subsequently prevents the NPs
the major trap of polymer chains between NCs takes place.66 After
aggregation.28 Moreover, the time of primary drying depends on
freezing, polymers and NCs merely have enough movement for
the depth of the vial and the formulation. The heat runs from the
involvement or crystal fusion.67 Accordingly, the freezing step is of
lowest to the highest part and sublimation is conversely carried out
much greater importance compared to sublimation step in order to
from highest to lowest part in the vial.19 Therefore, the process of
stop NPs from aggregating and save their ability to be re-dispersed.
improving the lyophilization cycle focuses on shortening the pri-
Significantly, the pace of fronts freezing (freezing rate), is identified
mary drying period that is the longest stage.51
primarily by gradient and may be a vital feature. Critical freezing
rate is described according to the plot of the particle dimensions of
Nanocrystals rebuilt NCs against the freezing speed. The rate of critical freezing
relies upon the empirical PS measuring details depended on API
Drug NCs are in fact nanoscopic crystals of the parent drug concentration.68 Quicker freezing rate yielded dry powders which
combined with excipients with sizes smaller than 1000 nm.52 The would be re-dispersed in water to produce the initial NPs.

Table 2
The Examples of Drug Nanocrystals Manufacturing Methods and Main Excipients Used for Freeze-Drying.

Compound Method Aid Excipients for Freeze-Drying Literature Reference


127
Albendazole HPH Mannitol
128
Danazol Media milling PVP, Tween® 80, mannitol, sucrose
129
Naproxen Media milling None
130
Oridonin HPH Mannitol
131
Ascorbylpalmitate HPH Trehalose
132
Fenofibrate Antisolvent percipitation Mannitol
133
Ursodeoxycholic acid High-pressure precipitation tandem homogenization Sucrose
134
Risperidone (Nanoprecipitation with probe sonication/ Trehalose dihydrate and sorbitol
nanoprecipitation with HPH/nanoprecipitation
3240 M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247

According to the results, a critical freezing rate, below which the external particles are kept out by molecules of water when HAR-
dried NCs cannot be re-dispersed, was identified based on the plot NCs having adequate time and also making them come close to
of the PS of reconstituted NCs versus freezing rate. When the each other and this finally leads to aggregation at low freezing rate
freezing of dispersion was carried out in a slower rate, the trap of (conservative and moderate state), and at the same time, the sta-
adsorbed polymers or fusion of API crystals, probably led to robust bilizers' activity proves to be ineffective due to the phase detach-
aggregations. On the other hand, when freezing rate was close to its ment.72 The best freezing rate may give suitable conditions to stop
critical rate, bimodal PS distribution in re-dispersed formulation NCs from aggregating or perhaps prevents the fusion of NCs
emerged showing singular particles along with the aggregates. It through the freezing step.69 In the case of dosage forms that are
was confirmed that the concentration of NPs has an impact on the dissolved in mouth, low freezing rate is highly acceptable because
critical freezing rate because when the concentration of NPs structures with giant pores are formed, which can be simply dis-
reduced from 11.5 to 6.1 wt %, the critical freezing rate decreased integrated in the presence of hydration. Although disintegration is
several times. When the dispersion concentration was 1.8 wt %, the fostered by giant pores, when we deal with the systems including
critical freezing rate was simply so low that was impossible to be suspended NCs, a dense cryo-concentrated stage through freezing
measured by the present equipments. Usually, reducing the con- may prompt aggregation.73 An annealing step has been conjointly
centration directly results in a rise in inter-particle distance and a noted for the mentioned advantages such as ice crystals' size
decrease in density of NPs which reduces the collision frequency of dispersal speeding up initial drying and reducing heterogeneity
NCs and decelerate the aggregation kinetics.69 Moreover, the sol- among samples.38 Furthermore, mannitol as the bulking agent was
vent/antisolvent ratio through freeze-drying stages (both freezing discovered to give a mix of amorphous and crystalline forms.74
and drying) is very important. It has been discovered that by uti- Particularly, when mannitol to drug ratios are low, crystallization
lizing a solution with lower water/tertiobutyl alcohol (TBA) ratio in might be optimized if an annealing stage is merged into the
the freezing step, quicker dissolution was resulted showing that the freezing stage.75 The kind of freezing influenced the disintegration
crystalline dispersions included smaller crystals. When crystalli- annealing and diminished the disintegration period whilst it was
zation happened through drying, employing solutions with higher deferred by progressive freezing. Strangely enough, the product's
water/TBA ratio led to the composition of finer solvent crystals. hardness or fracturability were merely influenced by freezing pa-
Accordingly, two variables of freezing rate and water/TBA ratio rameters (it might be noted that a product's fragility is indirectly
should be considered to modify the crystal size. When crystalliza- shown by fracturability and is measured according to the primary
tion happens in freezing phase, a higher nucleation rate might fracture's load value. More specifically, it indicates a product's
result from faster freezing or smaller water content of the solution. endurance toward fracture).73 Dissolution is postponed by sharp
As a result, a lot of nuclei are shaped leading to smaller crystals.70 At freezing in contrast to the progressive freezing and therefore, it
the same time, crystallization in drying phase might additionally appeared to receive a less considerable intensifying influence by
happen when the temperature increases higher than glass transi- the annealing. As a result of poor mechanical force, the act of
tion temperature of the maximally freeze-concentrated fraction dissolution occurred quicker by slow freezing and therefore, ice
(Tg') and below the eutectic temperature (Te). In such a case, what crystals increase led to a raise in the porosity within the freezing
is obtained is the rubbery state of the freeze-concentrated fraction phase.76
and crystallization will happen. The crystals enlargement is limited
to the interstitial areas extent between the solvent crystals. The Stabilizer Effect
researchers showed that the interstitial areas' dimensions between The size variations of NCs caused by lyophilization with PVP
the solvent crystals including the freeze-concentrated fraction may stabilizers showed separate affirmative impacts for PVP as a sta-
be altered by modifying the rate of freezing or the water/TBA ratio. bilizer. It meant that at low concentrations, PVP caused a growth in
When the rate of freezing was slow or there existed a high water/ the size of NCs. The PVP to drug ratio being significantly low, in-
TBA ratio, large solvent crystals formed having large inter-crystal creases the possibility of the particle's surface not being fully
spaces. However, smaller solvent crystals are going to be formed covered by adsorption.77 In meloxicam NCs, the areas of the
when the solutions were rapidly frozen (or when there exists a meloxicam particles which were not covered, might display ag-
minimum amount of water in the solution), leading to tiny inter- gregation capability and caused a moderate raise in the size. An
stitial areas. As the crystallization happens within the freeze- interactional impact between PVP as stabilizer and the concentra-
concentrated fraction, the ultimate dimensions of drug crystals tion of stabilizer demonstrated as a decrease in size which was
can be limited by the interstitial areas which exist between the resulted from high PVP content, confirms the previous hypothesis.
solvent crystals.70 The researchers defined a re-dispersibility index Accordingly, poloxamer in the varying concentrations produced an
(RDI) as D0/D where D0 and D represented the volume-weighed affirmative small impact on the size of crystals. It induced a
mean PS of the freshly prepared of ursodeoxycholic acid NC decrease in PS right down to 10.27% of the primary size. In contrast,
(UDCA-NC) and final PS after freeze drying, respectively. The RDI of a remarkable reduction in size was obtained from lyophilizing of
UDCA-NC depended on both the intensity of freezing and the type nanosuspensions with PEGs as stabilizer.76 As poloxamer led to
of stabilizers. The various freezing temperatures might lead to the lowering of PS in addition to protect meloxicam crystallinity, the
irrevocable aggregation of UDCA-NC throughout freezing step. As maximum reduction was observed in the formulations containing
an example, RDI 20 C, RDI 80 C and RDI 196 C of UDCA-NC stabi- 1% poloxamer and 5% mannitol prior to lyophilization.73 The type
lized by 10% w/v of PVP 30 K were 2.98, 9.01 and 14.16, respectively. and quantities of stabilizers in the same freezing method had
However, those of UDCA-NC which have been stabilized by 50% w/v dramatically significant diverse protection impacts on UDCA-NCs.
of PVP 30 K was 1.16, 5.32 and 9.65, accordingly. The mentioned Higher the concentration of stabilizers were, better re-
outcomes signify that the dispersibility of frozen UDCA-NC at the dispersibility was obtained. It suggested that the ability of being
standard freezing degree (meaning low freezing rate) was higher re-dispersed in UDCA-NCs prepared by 50% w/v concentration of
compared to the moderate and high temperatures (high freezing surfactants or polymer stabilizers turned out to be far superior to
rate).71 In another study it was declared that the re-dispersibility of the others with 25% and 10% w/v concentrations. Also, the results
harmine solid NCs (HAR-NCs) at the rigorous conditions (the showed that the surfactants (TPGS, Cremophor RH40 and Tween
highest freezing rate) was higher compared to the conservative and 80) proved to be more efficient in comparison with polymeric
moderate states. The probable explanation by authors was that the stabilizers (HPMC, NaCMC).71 The same results were obtained
M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247 3241

about harmine solid NCs (HAR-NCs). It was reported that re- polymer chains within the dry stage and prevent the polymer
dispersibility of HAR-NCs having high (50% w/v) concentration of stabilizer's steric barrier effect.83 It was additionally showed that at
surfactants or polymer stabilizers were largely superior to the 10% w/v concentration of TPGS, Tween 80 and HPMC as stabilizer,
others with moderate and low concentrations. Furthermore, the RDI of HAR-NCs in the presence of 400% w/v concentration of su-
quantity and type of stabilizers significantly influenced the re- crose, trehalose and sorbitol was closer to one in comparison with
dispersibility of HAR-NCs after freeze-drying.72 It may be reason- 100 and 200% of them. Furthermore, 100% w/v concentration of
able that HAR-NCs might approach to each other through crystal sucrose, glucose or sorbitol had a better function on HPMC stabi-
bridges when drying and they merge to form big agglomerates.78 lized HAR-NCs compared with alternative cryoprotectants.72
The polymeric stabilizers appeared to be adequately efficient to Moreover, NCs that have been made stable with Pluronic® F-68
guard the HAR-NCs during varying stresses throughout drying or Pluronic® F-127 indicated more growth in PS in the presence of
which might be due to their homogeneously adsorption onto the cryoprotectants. As a similar result, other researchers reported that
top layer of HAR-NCs and forming a steric barrier layer.72 the NCs containing solely SLS possessed smallest PS ratio.81

Cryoprotectant Type and Concentration Solvent Effect


The type and concentration of cryoprotectants compete for a Solvents that do not freeze and/or sublime fully using industrial
vital role in maintaining smart re-dispersibility options of NCs. To freeze dryers have been proved to be harder to be employed and
set the criterion of the freeze-drying cycle, initial evaluations of sometimes led to freeze-dried intermediates which were not
cryoprotectant and Tg values were carried out for all cryoprotectant satisfactory. By choosing solvent DMSO, the freeze-drying of gli-
solutions and a cryoprotectant blended with nanosuspensions benclamide NCs was unacceptable because the freeze-dried in-
too.79 Determining Tg for cryoprotectants separately and also in termediates happened to be greasy, yellow, and sticky and the
form of a mixture with nanosuspension indicated that effective solvent features of DMSO could describe that. DMSO is a solvent
cryoprotection was obtained by determining the smallest change in with high freezing point that is suitable for a decent freeze-drying
Tg values. In that sense, six types of sugars or polyols (glucose, method having really a low vapor pressure.84 For evaluating the
maltose, sucrose, lactose, mannitol, and sorbitol) (3%, w/v) in the solvent effects, various solvent mixtures were put into trial con-
role of lyophilization supports have been studied in preparation of sisting of DMSO (major solvent, reasonable solubility, low quality
azilsartan NCs. Sucrose was the foremost effective lyophilization freeze-drying outcomes) and TBA (secondary solvent, low solubil-
support and induced highest stability on lyophilization method.80 It ity, reasonable freeze-drying outcomes) in diverse solvent ratios to
was stated that by decreasing the mannitol's concentration to 5% enhance freeze-drying functionality. Characteristics such as high
and 2% w/v, the system became unstable. The results showed that at freezing point, high vapor pressure and low toxicity have made TBA
least 10% of mannitol and other protectants was required for pos- a suitable solvent for freeze-drying. Using TBA in the role of an
sessing a proper PS ratio.81 Moreover, it has been revealed that RDI organic co-solvent resulted in obtaining a satisfactory freeze-
of UDCA-NC when 10% w/v of cryoprotectants (like lactose and drying performance. A permeable and fluffy product was ob-
mannitol) was added to formulation was more than 2 whereas tained when TBA was used as the supplement solvent.85 The su-
adding 400% w/v of cryoprotectants (for instance sucrose, sorbitol) perior freeze-drying outcomes (fluffy cakes) were achieved with
showed RDI of less than 2.71 Moreover, it was observed that cryo- the DMSO: TBA ratios of 25:75 and 10:90. The amount of the
protectants sorbitol and sucrose had higher efficiency on RDI of organic solvents throughout the freeze-drying method has proved
HAR-NCs compared to other protectants. Also, the more the cryo- to be vital in changing the API's crystalline form to an amorphous
protectants' concentration increased, the higher protection under one. With DMSO:TBA 25:75, the drug crystal behavior changed
all process conditions was observed. These observations were from crystalline to amorphous, however too much TBA (for
supposed to be related to osmotic pressure effects. On the other example the DMSO: TBA 10:90 sample) caused to an increase in
words, these results could be explained by comparatively high crystalline behavior.84
concentrations of adsorbed polymers or sugars within the area
between both surfaces when they get closer to each other.72 A Nanoemulsions
previous study showed the importance of the treatments before
lyophilization on cellulose NCs (CNC) morphology. The researchers NEs known as oil-in-water (O/W) or water-in-oil (W/O) are
demonstrated that CNCs freeze-dried from 1% suspension had dispersions of two immiscible fluids being stable by use of an
smaller flakes compared with CNCs freeze-dried from 10.7% sus- acceptable surfactant.86 The mean droplet diameter appears to be
pension. However, sonication and flash freezing of the CNC sus- less than 500 nm.87 The small droplet size especially those less than
pensions resulted in fibrillary structures within the freeze-dried 100 nm provides them with a transparent or blurred look that
CNC morphology. This caused to a much higher degree of crystal- varies from milky white color related to large emulsions (which
linity. Consequently, the agglomerate sizes were decreased by micron-sized droplets take part in numerous light dispersions).88
dilution and/or increasing sonication time.82 NEs despite having a similar size differ from microemulsions
enormously in structural characteristics and the thermodynamic
Relationship Between Polymer Stabilizer and Cryoprotectants stability in the long time.89 The mentioned long-lasting physical
It has been considered that the ability of NCs for re-dispersion stability may directly be the result of small droplet size that pro-
may be related to a synergistic effect between polymer stabilizer hibits typical destabilization occurrences such as creaming, sedi-
and cryoprotectants. It has been found out that during the freezing, mentation and coalescence. Usually, brownian motion appears to
the cryoprotectants solution becomes freeze-concentrated and be adequately powerful to balance gravity or viscosity-evoked dy-
therefore produces a stable glass which is highly viscous. This namic instability.90 NEs are at risk of one major instability phe-
glassy state has minor mobility preventing NCs from fusion/ag- nomenon called Ostwald ripening. The mentioned phenomenon is
gregation and protects them against ice crystals. Lastly, the accountable for the increase in NEs' droplet size, by means of
hydrogen bonds of sugar reduce the interplays between the water inducing instability and it can be typically related to the system's
and UDCA-NCs and replace those of water in freeze-dried prod- aging. The event happens similarly in typical emulsions. Ostwald
uct.71,83 Moreover, the probable interplays between the head ripening could also be controlled via the modification of the
groups of sugars and polymeric stabilizers reduce the trap of droplets size and distribution. Using NEs is usually favored to
3242 M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247

microemulsions though they show more instability because the be beneficial, as it will not let the system freeze extraordinarily and
concentration of their surfactants are remarkably lower.91 Recently, would keep it from destruction. Furthermore, it maintains the rate
a study revealed that the droplet size of self-nanoemulsifying drug of freezing adequately high to ease secondary drying.96 The product
delivery systems (SNDDSs) increased when the freeze-drying temperature ought to be near to the Tg, since the enhancement of
method was applied. Nevertheless, this growth in the size was the temperature leads to the quicker procedure as some re-
tolerable as the droplet size remained below 200 nm but other searchers reported that a rise of one centigrade degree in the
physicochemical characteristics like polydispersity index (PDI), temperature of the product decreased the first drying time by
conduction and zeta potential, remained the same.92 The freeze thirteen fold.21 NEs probably have the shortest primary-drying time
drying of these formulations is influenced by the type and con- as a result of containing the biggest proportion of water which
centration of surfactants. When the unsaturation temperature is leads to a quicker mass transfer. The secondary-drying starts by a
low, the surfactant's acyl chains tend to be longer and when the slow-paced rise in the shelf temperature (0.1 or 0.15 C/min) since a
phase transition degree of the lipid is higher, additional drug loss is quick temperature change may result in the destruction of the
resulted.93 Although the emulsions, NEs and microemulsions might amorphous products due to having high unused humidity and
be precisely formed from the same ingredients, the microemulsions possessing a low Tg.21 Further, the instances with higher amount of
need a higher surfactant to oil ratio.94 Choosing the type and the particles dissolved in them, maintain smaller space within the dry
concentration of the cryoprotectant for NEs is vital. When the state. Consequently, higher temperature and an extended period is
cryoprotectant concentration is adequate, it can interrelate with required to get rid of the water.98 Table 3 summarizes the freeze-
the surface of the droplets and protect them through freeze-drying. dried NEs found in literature including the manufacturing
The most of cryoprotectants used in lyophilization of the emulsified method and main excipients used to optimize freeze-drying.
systems are disaccharides and saccharides. Amongst them, treha-
lose has appeared to have the highest efficiency. This superiority is
the result of its low hygroscopicity, ability to create inter-molecular Nanocapsules and Nanospheres
hydrogen bonds, small chemical reactivity as well as high Tg.95 The
perfect cryoprotectant concentration for emulsified systems fluc- NCPs consist of colloidal dispersions exhibiting a core-shell
tuates between 5% and 20% w/v.96 The study on freeze-dried structure which the core functions as a fluid reservoir for medi-
bufadienolides-loaded NEs (BU-NE) indicated that the protecting cines. The structure is usually composed of polymers, desirably
efficiency of maltose, trehalose and sucrose alone and in combi- decomposable, thick and stiff in the nanometric scale. The benefits
nation was remarkable whereas glucose wasn't effective. These of this kind of formation can be as follow; first, NCPs have high
excipients were the foremost effective at the identical concentra- medicine encapsulating efficiencies because of the maximized
tion of 20% w/v. Ironically, there was a rise in particle aggregation in solvability of drug within the NCP core and small polymer amount.
the presence of cryoprotectants with higher concentrations. In- Next, as the medicine is preserved in the NCP core and remains safe
stances including trehalose/sucrose and maltose/sucrose combi- from degradation. Furthermore, tissue irritation due to the burst
nations of varied compositions did not show to further improve effect at the administration site is low.99 Numerous strategies have
cryoprotective effect. The influence of various cryoprotectants been sometimes employed for creating this kind of core-shell
(glucose, fructose, mannitol, sucrose, lactose, trehalose and construction on a nanometric scale including in-situ polymeriza-
maltose) on bufadienolides-loaded submicron-emulsions (BU-SE) tion at the NE droplet interface, NCP generation by use of pre-
has been investigated. The existence of an exact quantity of formed polymer and also generating of lipid NCPs. Lipophilic and
amorphous disaccharides in lyophilization proved to be an essen- hydrophilic parts of the NCPs are distinguishable provided that the
tial issue for rehydrated BU-SE. The rehydrated BU-SE including NCPs are dispersed in water.100
mannitol, glucose, fructose, and lactose did not reveal a Tindall On the other hand, a polymeric NSP could be described as a
effect. The lyophilization results suggested that the best cryopro- matrix kind, firm colloidal particle which in drugs are solved,
tectants for BU-NE and BU-SE have been maltose and trehalose at trapped, or enclosed with the chemicals bound or adsorbed onto
an identical concentration of 20% w/v.97 Adding cryoprotectant to the polymer matrix. The mentioned particles are generally bigger
the NEs was accomplished after the completion of the process. The compared to micelles which have diameters ranging from one
results demonstrated differences in NE features such as droplet size hundred to two hundred nanometer and usually show significantly
and drug loading capacity, due to adding cryoprotectant at various higher polydispersity.101
stages. Adding cryoprotectants in the step of preparing NEs led to Freeze-drying is thought-about as a decent methodology which
an increase in droplet size and polydispersity. However, when the preserves the unification of colloidal dispersions. Thus, the freeze-
researchers tried to add the cryoprotectants in the last formulation, drying of NCPs and NSPs which possess slim and frangible structure
it was troublesome to dissolve a number of the sugars like mannitol barely resistant to the mechanical force of the process has special
and lactose. Also, a rise in the droplet size and drug leaking would considerations. Therefore, it is important to examine the features
be resulted in the case of using ultrasound.97 which may have an effect on the NCPs and NSPs stability
Generally, the freezing states ought to be cautiously identified. throughout the various stages of lyophilization. Table 4 summarizes
In slow freezing, as the water will diffuse in a slow rate, the osmotic the freeze-dried NCPs and NSPs found in literature including the
pressure is decreased stopping the outpouring of the droplets.98 manufacturing method and main excipients used to optimize
Nevertheless, the application of average freezing rates may well freeze-drying.

Table 3
The Examples of Drug Nanoemulsions Manufacturing Methods and Main Excipients Used for Freeze-Drying.

Compound Method Aid Excipients for Freeze-Drying Literature Reference


135
Febuxostat Self Nanoemulsifying Drug Delivery Systems Trehalose
136
Ginkgo biloba Emulsion solvent evaporation Hydroxy beta cyclo dexterin
97
Bufadienolides Ultrasonic-high-pressure homogenization Maltose and trehalose
92
Adefovir dipivoxil Self Nanoemulsifying Drug Delivery Systems D-mannitol
M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247 3243

Table 4
The Examples of Drug Nanocapsules and Nanospheres Manufacturing Methods and Main Excipients Used for Freeze-Drying.

Compound Method Aid Excipients for Freeze-Drying Literature Reference


137
Vitamin E Nanoprecipitation Sucrose
138
Chitosan containing Docetaxel Solvent displacement Trehalose
139
Capsicum Emulsionediffusion Pluronic F68
108
Itraconazole Emulsion solvent evaporation Sucrose, Glucose, Lactose, Trehalose, Mannitol
140
Mitoxantrone Nanoprecipitation/solvent diffusion Lactose
141
Poly(ester-anhydride) Water-in-oil-in-water double emulsion Trehalose
142
Methacrylic acid Solution/precipitation polymerization Pluronic F68
143
Ketorolac tromethamine Microreactor technique Mannitol

Freeze-Drying Cycle Effect was 1.15 which is acceptable. Also, the procedure conducted
About NCPs, diverse dispersions were discovered at completely at 15  C, 10  C, and 5  C showed that a rise in the ratio of NCPs
distinct places within the freeze-dried bulk sample, and this non- PDI pre and post annealing was 1.68, 1.92, and 2.31, accordingly.106
homogenity has been obsessed with the cooling program utilized
within the process. As an instance, when gelatin NCPs mixture's Formulation Effect
temperature was quickly reduced and it was frozen, bottom layers' The cryoprotectant nature and concentration have vital roles in
dispersibility was lower compared to the layers at the top, and this the lyophilization process. Disaccharides which are stable chemical
tendency appeared more obvious at the external portions. In additives proved to be beneficial excipients able to vitrify simply
another example, when freezing of the sample suspensions took throughout the freezing stage.36 The characteristic disaccharides to
place at a continual slow cooling rate, dispersions had a very small make a glassy matrix within which the NPs are fixed, prevent the
nonhomogenity within the dried bulk sample. In addition, in particles from aggregating.107 Disaccharides such as sucrose and
comparison with fast cooling it had a better dispersibility. It was trehalose appeared to be consistent with polybutyl cyanoacrylate
discovered that there is a mutual relationship between gelatin (PBCA) and thus functioned as perfect cryoprotectants for PBCA-
NCPs' dispersibility and the freezing front speed, and therefore with NSP loading itraconazole. Glucose and lactose failed to stop itra-
the viscosity at or around the freezing front. It has been recom- conazole leakage when reconstitution.108 Same as mannitol, lactose
mended that the forming of gel is beneficial to prevent undesirable tends to crystallize more even within the freeze-drying cake in
denaturing and to provide better dispersion within the dried ma- comparison with alternative disaccharides.36 This will result in
trix. Nevertheless, slow dehydrating might result in extra stress due phase division and therefore the NPs complicated structure can be
to the development of enormous ice crystals resulting in low dis- damaged. As the researchers reported, the freeze-dried cake con-
persibility within the ultimate dried product.102 The lower freezing taining 10% glucose fell down and it was rather hard to suspend the
temperatures were additionally effective for keeping the initial size lyophilized cake again. It seems that in this case, the collapse
and morphology of NPs. The mean PS of freeze-dried fish oil NCPs temperature (CT) went over and caused to structural collapse
was considerably raised by reducing the freezing temperature. within the dried matrix and itraconazole leakage when reconsti-
Therefore, to optimize the lyophilization procedure, higher freezing tution. Moreover, it may well be revealed that the cryopreservation
temperature was the most favorable state to keep up the fish oil quality of disaccharides like sucrose and trehalose is related to their
quantity within the particles and delay oxidation.103 It has been concentration inside the nano-formulation. A cryoprotectant con-
demonstrated that the sublimation rate increased and the hetero- centration lower than 10% was not enough to preserve the original
geneity of the sample decreased by annealing.104 Annealing had an itraconazole loading and construct a preventing vitrification layer
effect on initial drying by speeding up the sublimation rate with no around the PBCA-NSPs. For the long term, the researchers have
change in the size of NCPs. The sublimation rate of NCPs utilizing used trehalose as choice cryoprotectant because it is highly stable
sucrose and PVP as protectants showed the sublimation rate of in conditions with high temperature and humidity in comparison
87.9 mg/h for sucrose and 70 mg/h for PVP. However, annealing to sucrose. This feature is often due to the higher Tg of dried
produced the best sublimation rates. Using sucrose, the sublima- trehalose matrix when adsorbing vapor.108 Studies revealed that
tion rate increased from 86.7 to 124.8 mg/h, while about PVP, the the dominant destabilization cause in freeze-drying of NSPs was
sublimation rate rose from 77 to 83.9 mg/h when the annealing ineffective surface modifications and significant drug desorption.
temperature went up from 20 to 10  C.50 These findings indi- Therefore, the hydrophilic surfactants adsorbed at NSPs surface had
cated that accelerating the sublimation rate relied on the annealing a great impact on cryopreservation. Replacement of the normally
temperature. employed non-ionic surfactant pluronic F68 by the anionic sur-
The impact of annealing on secondary drying depends on the factant sodium deoxycholate led to a very successful stabilization of
type of cryoprotectant employed. Studies showed that annealing of itraconazole-loaded NSPs without desorbing the drug, albeit when
sucrose solution slowed down the secondary drying kinetic while 10% sucrose was present and not glucose. Pluronic F68 might
there was not any impacts using PVP. Sucrose seems to be slightly crystallize through freezing leading to surface alternations and
less hygroscopic compared to PVP. This sort of dissimilarity in hy- drug desorption whereas this might not happen with sodium
groscopicity could be responsible to the difference between the deoxycholate.109 It may be observed that if the stabilizer's con-
secondary drying kinetics of the compounds.105 Searle's, Carpenter centration is raised, a decrease in the size and polydispersity of
and Randolph indicated that following an annealing process, the NSPs is resulted. The researchers showed that when the PVA con-
drying rate might rise up to 3.5 fold and only 30 min of annealing centration as stabilizer was raised from 0.62 to 5% w/v, the ratios of
could improve the rate significantly.104 It has been found that by NCPs size after and before lyophilization (SF/SI) decreased from 4 to
annealing of ovalbumin loaded carboxymethyl chitosan NCPs 1 indicating that the initial size was remained unchanged. The PVA
at 15  C, the NCPs sizes did not considerably change and therefore, concentration of 2.5% w/v was the lowest of which was needed for
the ratio of NCPs sizes pre and post annealing was close to one. stabilizing the NCPs when freezing. Similarly, about poly (epsilon-
Although, when the procedure was carried out at 25  C, NP size caprolactone) (PCL) NCPs, 2.5% w/v of sucrose was the lowest
ratio was raised, the ratio of the NCPs PDI pre and post annealing concentration required to enhance NCPs stability. Furthermore, the
3244 M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247

increase of PCL concentration induced a rise of size though the stabilizing impact of trehalose. Accordingly, the lyophilization of
calculated envelope thickness was in the accepted range. The trial docetaxel-loaded chitosan NCPs without poloxamer revealed that
of various concentrations of PCL demonstrated that the concen- the freeze-drying of drug-loaded NCPs might take place with the
tration of PCL did not enhance the NCPs stability when freezing identical quantities of trehalose and the resulting freeze-dried
probably due to the increase of NCPs size. The freeze-thawing product might be properly reconstructed with no loss of for-
stability of PCL NCPs was obtained from two oils (miglyol and sil- mulation's characteristics.111 Another team of researchers studied
icon oil). Consequently, preserving the enclosed oil in the liquid the effect of different concentrations of Hyaluronan NCPs (0.5 and
condition did not increase the stability of NCPs in the freezing step. 1% w/v) and trehalose (5% and 10% w/v) using benzalkonium
Thus, it may be assumed that the stabilizer proportion utilized in chloride as the surfactant. The results showed that at rather high
the formulation and also cryoprotectant are the foremost vital el- concentrations of NCPs (1% w/v), it was likely to attain an
ements that affect the stability in the freezing step.50 These fragile appropriate re-suspension of the freeze-dried product without
particles may be destabilized in the freezing or the dehydration significant change in the size of the NCPs.112
steps owing to the crystallization of for instance mannitol. Under
storage time, NCPs freeze-dried in the presence of sucrose and Cocrystals/Nanococrystals
glucose may well be aggregated if exposed to high temperatures
and moderate humidities. The mentioned state might begin the Cocrystals are in fact “homogenous” (single phase) crystalline
crystallization of amorphous lyoprotectants inducing aggregation forms created from two or more compounds possessing a distinct
of NCPs. Therefore, freeze-dried NCPs ought to be kept at a tem- stoichiometric quantitative ratio wherever the positioning within
perature lower than the glass transition temperature of the the crystal lattice does not rely on ionic bonds.113 Cocrystal
formulation to keep up the glassy condition of lyoprotectant and to structure relies on noncovalent interplay of the API and also the
stop aggregation of NCPs. These findings were related to the NCPs components known as conformer. The involving interactions are
which have been freeze-dried with PVP and preserved their sta- intermolecular ones like hydrogen bonding, van der Waals con-
bility for only six months of being stored in stress conditions.29 tact forces, p stacking, static interaction and halogen bonding
It is worthy to mention that the characteristics of the bio- among stoichiometric quantities of assorted molecules.114 Coc-
polymers and crosslinking method have made the direct freeze- rystallization strategies are classified based on the employment
drying of NCPs possible without the need to use common poly- (or not) of solvent, so the cocrystallization methods are catego-
ols cryoprotectants. The biopolymers having global structure will rized as solvent-based or solvent-free. Solvent-based strategies
stick onto the surface of NCPs with high affinity via different are the most routine and accepted ones particularly for laboratory
crosslinking methods forming namely a “rigid wall” preserving the small-sized utilization. The mentioned strategies can sometimes
lipid core. As it was indicated in the preparation of NE-templated be unsafe due to using considerable quantities of hazardous
shell-crosslinked NCPs of fenofibrate, when the aqueous solvents.115
biopolymer was freeze-dried, these biopolymers were glassified Freeze drying may be a technique usually resulting to amor-
forming a lyocake having a suitable quality without showing the phous substances though it can be sometimes appropriate for
indications of collapse. These observations propose that the free producing cocrystals too. For producing cocrystals, certain molar
biopolymers themselves will work in the role of cryoprotectants quantities of the compounds are dissolved in a suitable solvent. The
preserving the product against freezing and drying stresses.110 The solvent choice is performed according to instrument chemical
restoration of the primary characteristics of chitosan NCPs on compatibility and solvent capability for dissolving the API and
lyophilization and reconstruction was different based on the coformer efficiently. The resulting solution is subjected to a quick
concentration of NCPs (0.25, 0.5 and 1% w/v) and cryoprotectant (5 freezing and also a high vacuum which results in subliming the
and 10% w/v). The most effective outcomes were achieved for a solvent. A low-density powder is created, typically in amorphous
NCP concentration of 0.25% w/v and trehalose concentration of form. If the glass transition temperature of the product is lower
10% w/v. The freeze drying of chitosanepoloxamer NCPs in pres- than ambient temperature, amorphous forms are prone to form.
ence of 10% w/v of trehalose indicated a 50e100 nm rise in the However, cocrystallization with this methodology does not have
size of NCPs which can be attributed to higher crystallinity of any kinetic obstacles to produce crystalline forms.116 For instance,
glassy matrix structure in the presence of poloxamer. This with dissolving theophylline and various coformers in water or
augmented crystallinity might be the reason of the lower tertiobutanol as solvent, the cocrystals of many forms were

Table 5
Key Considerations in Freeze Drying of Different Classes of Nanoparticles Including Formulation Parameters and Optimum Freeze-Drying Conditions.

Nanoparticles Classifications Most Influential Formulation Parameters Optimum Freeze-Drying Conditions

Nanosuspensions Stabilizer structure and conc. Slow/fast freezing rate


Cryo/lyoprotectant type and conc. Short primary drying Period
NPs conc.
Nanocrystals Cryo/lyoprotectant Tg and conc. Fast freezing rate (most often)
Stabilizer-cryoprotectant interaction Short primary drying period
Solvent characteristics
Solvent to anti-solvent ratio
NCs conc.
Nanoemulsions Stabilizer structure and conc. Moderate freezing rate
Cryo/lyoprotectant conc. Higher temperature in freezing/drying periods
Extended freezing/drying periods
Short primary drying period
Nanocapsules/Nanospheres Stabilizer structure and conc. High/low freezing temperature
Shell crosslinking degree Application of annealing
Cryo/lyoprotectant type and Conc. Short primary drying period
NCPs conc
M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247 3245

obtained including completely unique solid solution of theo- Conflicts of Interest


phylline:caffeine and a possible new cocrystal kind of theophylli-
ne:oxalic acid. X-ray powder diffraction (XRPD) and DSC analyses The authors declare no conflict of interest, financial or
showed the existence of a pentahydrate theophylline:oxalic acid otherwise.
2:1 cocrystal for the freeze-dried instances. About the theophylli-
ne:caffeine, the analyses demonstrated the same paradigm; how- Acknowledgements
ever, lack of an amorphous form that would be ascribed to the
single component phase of theophylline indicated a solid solution The authors would like to thank pharmaceutics laboratories of
of the two constituents. Additional examinations demonstrated the Shiraz School of Pharmacy for providing detailed information about
forming of single NeH bond between the two molecules.117 How- freeze-drying instrument.
ever, the mentioned methods additionally had a number of disad-
vantages. They needed adequate API and coformer solubility in References
appropriate solvents, particularly water and t-butanol. In addition,
when solidifying is fast, amorphous solids or crystals with less 1. Shi J, Votruba AR, Farokhzad OC, Langer R. Nanotechnology in drug delivery
stability might be produced. and tissue engineering: from discovery to applications. Nano Lett. 2010;10(9):
3223-3230.
Freeze-drying is appropriate for the sterile producing of solid 2. Sung JC, Pulliam BL, Edwards DA. Nanoparticles for drug delivery to the lungs.
formulations for injection.118 Main advantages of cocrystal Trends Biotechnol. 2007;25(12):563-570.
manufacturing by freeze-drying include being single step and 3. Gao L, Zhang D, Chen M. Drug nanocrystals for the formulation of poorly
soluble drugs and its application as a potential drug delivery system.
continuous process, easy scale up, and prevention of single- J Nanoparticle Res. 2008;10(5):845-862.
compound crystals formation.116 Freeze-drying might be helpful 4. Desai MP, Labhasetwar V, Walter E, Levy RJ, Amidon GL. The mechanism of
in many phases of cocrystal formation from primary cocrystal uptake of biodegradable microparticles in Caco-2 cells is size dependent.
Pharm Res. 1997;14(11):1568-1573.
screening and examination of cocrystal polymorphism to prepa- 5. Fonte P, Araújo F, Reis S, Sarmento B. Oral insulin delivery: how far are we?
ration of cocrystals in industrial scale.117 J Diabetes Sci Technol. 2013;7(2):520-531.
Although the co-crystal and nanocrystal technologies have been 6. Courrier H, Butz N, Vandamme TF. Pulmonary drug delivery systems: recent
developments and prospects. Crit Rev Ther Drug Carrier Syst. 2002;19(4-5).
widely used in the development of pharmaceuticals, but their 7. Lemoine D, Francois C, Kedzierewicz F, Preat V, Hoffman M, Maincent P.
synergistic effect has not been well investigated. Nanococrystals, Stability study of nanoparticles of poly (e-caprolactone), poly (d, l-lactide) and
which combine benefits of both technologies can be used to solve poly (d, l-lactide-co-glycolide). Biomaterials. 1996;17(22):2191-2197.
8. Wu L, Zhang J, Watanabe W. Physical and chemical stability of drug nano-
the problem of water-insoluble drugs. Therefore, similar to nano-
particles. Adv Drug Deliv Rev. 2011;63(6):456-469.
crystals, lyophilization can be used to produce solid forms of 9. Chacon M, Molpeceres J, Berges L, Guzman M, Aberturas M. Stability and
nanocorystals. Athough there aren't many studies on these systems, freeze-drying of cyclosporine loaded poly (D, L lactideeglycolide) carriers. Eur
it seems that the same considerations as nanocrystals could be J Pharm Sci. 1999;8(2):99-107.
10. Auvillain M, Cave  G, Fessi H, Devissaguet JP. Lyophilisation de vecteurs col-
imagined for nanococrystals too. loïdaux submicroniques. STP Pharma. 1989;5(11):738-744.
Table 5 summarizes the most influential parameters related to 11. Katas H, Hussain Z, Rahman SA. Storage stabilisation of albumin-loaded chi-
NPs formulation and the most optimum freeze drying conditions in tosan nanoparticles by lyoprotectants. Trop J Pharm Res. 2013;12(2):135-142.
12. Boge L, et al, V€ astberg A, Umerska A. Freeze-dried and re-hydrated liquid
which the best results would be achieved. crystalline nanoparticles stabilized with disaccharides for drug-delivery of the
plectasin derivative AP114 antimicrobial peptide. J Colloid Interface Sci.
2018;522:126-135.
13. Chung N-O, Lee MK, Lee J. Mechanism of freeze-drying drug nanosuspensions.
Conclusion Int J Pharm. 2012;437(1-2):42-50.
14. Bozdag S, Dillen K, Vandervoort J, Ludwig A. The effect of freeze-drying with
The lyophilization procedure is of great importance for different cryoprotectants and gamma-irradiation sterilization on the charac-
teristics of ciprofloxacin HCl-loaded poly (D, L-lactide-glycolide) nano-
improving the long-term stability of NPs. Therefore, the therapeutic
particles. J Pharm Pharmacol. 2005;57(6):699-707.
goals and specifications of the drug must be considered for 15. Lu X, Pikal MJ. Freeze-drying of mannitoletrehaloseesodium chloride-based
lyophilization. To achieve a good product, there exist numerous formulations: the impact of annealing on dry layer resistance to mass transfer
and cake structure. Pharm Dev Technol. 2004;9(1):85-95.
factors in NPs formulation and also freeze-drying process which
16. Wong JJL, Yu H, Hadinoto K. Examining practical feasibility of amorphous
need to be optimized. The most influential factors include the type curcumin-chitosan nanoparticle complex as solubility enhancement strategy
and concentration of cryoprotectant and stabilizer, the relationship of curcumin: scaled-up production, dry powder transformation, and long-
between protectant and stabilizer, the concentration of NPs, term physical stability. Colloid Surface Physicochem Eng Aspect. 2018;537:36-
43.
freezing, annealing, and drying rate. Without these considerations, 17. Fonte P, Reis S, Sarmento B. Facts and evidences on the lyophilization of
the resulting freeze-dried product may not be able to reproduce the polymeric nanoparticles for drug delivery. J Control Release. 2016;225:75-86.
primary NPs dispersions regarding their PS and PDI. However, the 18. Franks F. Freeze drying: from empiricism to predictability. Cryo Lett. 1990;11:
93-110.
freezing of NPs encounters the challenges that need further 19. Pikal M, Shah S, Roy M, Putman R. The secondary drying stage of freeze
research to overcome. One of these is the scale-up considerations drying: drying kinetics as a function of temperature and chamber pressure. Int
and physical stability of lyophilized NPs to reach to the pharma- J Pharm. 1990;60(3):203-207.
20. Williams N, Polli G. The lyophilization of pharmaceuticals: a literature review.
ceutical market. In this regard, the theoretical mathematical PDA J Pharm Sci Technol. 1984;38(2):48-60.
models can be employed to design, control, optimize and scale-up 21. Tang XC, Pikal MJ. Design of freeze-drying processes for pharmaceuticals:
different phases of freeze-drying processes. Also, lyophilization can practical advice. Pharm Res. 2004;21(2):191-200.
22. Bejrapha P, Surassmo S, Choi MJ, Nakagawa K, Min S-G. Studies on the role of
be used in the future to process the new developing types of NPs gelatin as a cryo-and lyo-protectant in the stability of capsicum oleoresin
like nanotubes, nanofibers, and nanoaggregates in industrial scale nanocapsules in gelatin matrix. J Food Eng. 2011;105(2):320-331.
provided that these delivery systems could meet the safety re- 23. Sadikoglu H, Ozdemir M, Seker M. Freeze-drying of pharmaceutical products:
research and development needs. Dry Technol. 2006;24(7):849-861.
quirements for using in humans.
24. Andreani T, et al, Kiill CP, de Souza ALR. Effect of cryoprotectants on the
reconstitution of silica nanoparticles produced by solegel technology. J Therm
Anal Calorim. 2015;120(1):1001-1007.
Ethics Approval and Consesent for Publication 25. Allison SD, dC Molina M, Anchordoquy TJ. Stabilization of lipid/DNA com-
plexes during the freezing step of the lyophilization process: the particle
isolation hypothesis. Biochim Biophys Acta Biomembr. 2000;1468(1-2):127-
Not applicable. 138.
3246 M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247

26. Crowe LM, Reid DS, Crowe JH. Is trehalose special for preserving dry bio- 54. Merisko-Liversidge E, Liversidge GG, Cooper ER. Nanosizing: a formulation
materials? Biophys J. 1996;71(4):2087-2093. approach for poorly-water-soluble compounds. Eur J Pharm Sci. 2003;18(2):
27. Almalik A, Alradwan I, Kalam MA, Alshamsan A. Effect of cryoprotection on 113-120.
particle size stability and preservation of chitosan nanoparticles with and 55. Keck CM, Müller RH. Biopharmaceutics, drug nanocrystals of poorly soluble
without hyaluronate or alginate coating. Saudi Pharm J. 2017;25(6):861-867. drugs produced by high pressure homogenisation. Eur J Pharm Biopharm.
28. Lee MK, Kim MY, Kim S, Lee J. Cryoprotectants for freeze drying of drug nano- 2006;62(1):3-16.
suspensions: effect of freezing rate. J Pharm Sci. 2009;98(12):4808-4817. 56. Sarkari M, Brown J, Chen X, Swinnea S, Williams III RO, Johnston KP. Enhanced
29. Abdelwahed W, Degobert G, Fessi H. Investigation of nanocapsules stabili- drug dissolution using evaporative precipitation into aqueous solution. Int J
zation by amorphous excipients during freeze-drying and storage. Eur J Pharm Pharm. 2002;243(1e2):17-31.
Biopharm. 2006;63(2):87-94. 57. Shekunov BY, Chattopadhyay P, Seitzinger J, Huff R. Nanoparticles of poorly
30. Kamiya S, Nozawa Y, Miyagishima A, Kurita T, Sadzuka Y, Sonobe T. Physical water-soluble drugs prepared by supercritical fluid extraction of emulsions.
characteristics of freeze-dried griseofulvin-lipids nanoparticles. Chem Pharm Pharm Res. 2006;23(1):196-204.
Bull. 2006;54(2):181-184. 58. Kipp J. The role of solid nanoparticle technology in the parenteral delivery of
31. Umerska A, Paluch KJ, Santos-Martinez MJ, Corrigan OI, Medina C, Tajber L. poorly water-soluble drugs. Int J Pharm. 2004;284(1-2):109-122.
Freeze drying of polyelectrolyte complex nanoparticles: effect of nanoparticle 59. Blagden N, de Matas M, Gavan PT, York P. Crystal engineering of active
composition and cryoprotectant selection. Int J Pharm. 2018;552(1e2):27-38. pharmaceutical ingredients to improve solubility and dissolution rates. Adv
32. Gokce Y, Cengiz B, Yildiz N, Calimli A, Aktas Z. Ultrasonication of chitosan Drug Deliv Rev. 2007;59(7):617-630.
nanoparticle suspension: influence on particle size. Colloid Surface Phys- 60. Fischer HC, Chan WC. Nanotoxicity: the growing need for in vivo study. Curr
icochem Eng Aspect. 2014;462:75-81. Opin Biotechnol. 2007;18(6):565-571.
33. Müller RH, Gohla S, Keck CM. State of the art of nanocrystalsespecial features, 61. Rabinow BE. Nanosuspensions in drug delivery. Nat Rev Drug Discov.
production, nanotoxicology aspects and intracellular delivery. Eur J Pharm 2004;3(9):785.
Biopharm. 2011;78(1):1-9. 62. Van Eerdenbrugh B, Van den Mooter G, Augustijns P. Top-down production of
34. Du J, et al, Li X, Zhao H. Nanosuspensions of poorly water-soluble drugs drug nanocrystals: nanosuspension stabilization, miniaturization and trans-
prepared by bottom-up technologies. Int J Pharm. 2015;495(2):738-749. formation into solid products. Int J Pharm. 2008;364(1):64-75.
35. Konan YN, Gurny R, Alle mann E. Preparation and characterization of sterile 63. Patravale V, Date AA, Kulkarni RM. Nanosuspensions: a promising drug de-
and freeze-dried sub-200 nm nanoparticles. Int J Pharm. 2002;233(1-2):239- livery strategy. J Pharm Pharmacol. 2004;56(7):827-840.
252. 64. de Waard H, Grasmeijer N, Hinrichs WL, Eissens AC, Pfaffenbach PP, Frijlink
36. Franks F. Freeze-drying of bioproducts: putting principles into practice. Eur J HW. Preparation of drug nanocrystals by controlled crystallization: applica-
Pharm Biopharm. 1998;45(3):221-229. tion of a 3-way nozzle to prevent premature crystallization for large scale
37. De Jaeghere F, Allemann E, Leroux JC, et al. Formulation and lyoprotection of production. Eur J Pharm Sci. 2009;38(3):224-229.
poly (lactic acid-co-ethylene oxide) nanoparticles: influence on physical sta- 65. Sinha B, Müller RH, Mo €schwitzer JP. Bottom-up approaches for preparing
bility and in vitro cell uptake. Pharm Res. 1999;16(6):859-866. drug nanocrystals: formulations and factors affecting particle size. Int J Pharm.
38. Abdelwahed W, Degobert G, Stainmesse S, Fessi H. Freeze-drying of nano- 2013;453(1):126-141.
particles: formulation, process and storage considerations. Adv Drug Deliv Rev. 66. Lee J. Drug nano-and microparticles processed into solid dosage forms:
2006;58(15):1688-1713. Physical Properties. J Pharm Sci. 2003;92(10):2057-2068.
39. Sahoo SK, Panyam J, Prabha S, Labhasetwar V. Residual polyvinyl alcohol 67. Gedde UW. Polymer Physics. Springer Science & Business Media; 2013.
associated with poly (D, L-lactide-co-glycolide) nanoparticles affects their 68. Lee J, Cheng Y. Critical freezing rate in freeze drying nanocrystal dispersions.
physical properties and cellular uptake. J Control Release. 2002;82(1):105-114. J Control Release. 2006;111(1-2):185-192.
40. Galindo-Rodriguez S, Allemann E, Fessi H, Doelker E. Physicochemical pa- 69. Lee J, Cheng Y. Critical freezing rate in freeze drying nanocrystal dispersions.
rameters associated with nanoparticle formation in the salting-out, emulsi- J Control Release. 2006;111(1-2):185-192.
fication-diffusion, and nanoprecipitation methods. Pharm Res. 2004;21(8): 70. De Waard H, Hinrichs W, Frijlink H. A novel bottomeup process to produce
1428-1439. drug nanocrystals: controlled crystallization during freeze-drying. J Control
41. Molpeceres J, Aberturas M, Chacon M, Berges L, Guzman M. Stability of Release. 2008;128(2):179-183.
cyclosporine-loaded poly-X-caprolactone nanoparticles. J Microencapsul. 71. Ma Y-Q, Zhang ZZ, Li G, Zhang J, Xiao HY, Li XF. Solidification drug nano-
1997;14(6):777-787. suspensions into nanocrystals by freeze-drying: a case study with urso-
42. Beirowski J, Inghelbrecht S, Arien A, van Assche I, Gieseler H. Stabilization of deoxycholic acid. Pharm Dev Technol. 2016;21(2):180-188.
nanosuspensions during freeze-drying: the role of vitrification (Part 1). In: 72. Yue P, Wang C, Dan J, Liu W, Wu Z, Yang M. The importance of solidification
Proceedings of the Seventh World Meeting on Pharmaceutics. Biopharmaceutics stress on the redispersibility of solid nanocrystals loaded with harmine. Int J
and Pharmaceutical Technology; 2010. Pharm. 2015;480(1e2):107-115.
43. Beirowski J, Inghelbrecht S, Arien A, Gieseler H. Freeze-drying of nano- 73. Iurian S, Bogdan C, Tomuța  L, et al. Development of oral lyophilisates con-
suspensions, 1: freezing rate versus formulation design as critical factors to taining meloxicam nanocrystals using QbD approach. Eur J Pharm Sci.
preserve the original particle size distribution. J Pharm Sci. 2011;100(5):1958- 2017;104:356-365.
1968. 74. Torrado S, Torrado S. Characterization of physical state of mannitol after
44. Beirowski J, Inghelbrecht S, Arien A, Gieseler H. Freeze drying of nano- freeze-drying: effect of acetylsalicylic acid as a second crystalline cosolute.
suspensions, 2: the role of the critical formulation temperature on stability of Chem Pharm Bull. 2002;50(5):567-570.
drug nanosuspensions and its practical implication on process design. J Pharm 75. Mehta M, Bhardwaj SP, Suryanarayanan R. Controlling the physical form of
Sci. 2011;100(10):4471-4481. mannitol in freeze-dried systems. Eur J Pharm Biopharm. 2013;85(2):207-213.
45. Wang L, et al, Ma Y, Gu Y. Cryoprotectant choice and analyses of freeze-drying 76. Iurian S, Tomuta I, Bogdan C, et al. Defining the design space for freeze-dried
drug suspension of nanoparticles with functional stabilisers. J Microencapsul. orodispersible tablets with meloxicam. Drug Dev Ind Pharm. 2016;42(12):
2018;35(3):241-248. 1977-1989.
46. De Jaeghere F, Allemann E, Feijen J, Kissel T, Doelker E, Gurny R. Freeze-drying 77. Wang Y, Zheng Y, Zhang L, Wang Q, Zhang D. Stability of nanosuspensions in
and lyopreservation of diblock and triblock poly (lactic acid)epoly (ethylene drug delivery. J Control Release. 2013;172(3):1126-1141.
oxide)(PLAePEO) copolymer nanoparticles. Pharm Dev Technol. 2000;5(4): 78. Wang B, Zhang W, Zhang W, Mujumdar AS, Huang L. Progress in drying
473-483. technology for nanomaterials. Dry Technol. 2005;23(1e2):7-32.
47. Franks F. Freeze-drying: from empiricism to predictability. The significance of 79. Saez A, Guzman M, Molpeceres J, Aberturas M. Freeze-drying of poly-
glass transitions. Dev Biol Stand. 1992;74:9-18. discussion 19. caprolactone and poly (D, L-lactic-glycolic) nanoparticles induce minor par-
48. Kasper JC, Friess W. The freezing step in lyophilization: physico-chemical ticle size changes affecting the oral pharmacokinetics of loaded drugs. Eur J
fundamentals, freezing methods and consequences on process performance Pharm Biopharm. 2000;50(3):379-387.
and quality attributes of biopharmaceuticals. Eur J Pharm Biopharm. 80. Ma J, Yang Y, Sun Y, Sun J. Optimization, Characterization and in Vitro/vivo
2011;78(2):248-263. Evaluation of Azilsartan. .; 2016.
49. Cui Z, Hsu C-H, Mumper RJ. Physical characterization and macrophage cell 81. Thakkar S, Sharma D, Misra M. Comparative evaluation of electrospraying and
uptake of mannan-coated nanoparticles. Drug Dev Ind Pharm. 2003;29(6): lyophilization techniques on solid state properties of Erlotinib nanocrystals:
689-700. assessment of In-vitro cytotoxicity. Eur J Pharm Sci. 2018;111:257-269.
50. Abdelwahed W, Degobert G, Fessi H. A pilot study of freeze drying of poly 82. Han J, Zhou C, Wu Y, Liu F, Wu Q. Self-assembling behavior of cellulose
(epsilon-caprolactone) nanocapsules stabilized by poly (vinyl alcohol): nanoparticles during freeze-drying: effect of suspension concentration, par-
formulation and process optimization. Int J Pharm. 2006;309(1-2):178- ticle size, crystal structure, and surface charge. Biomacromolecules.
188. 2013;14(5):1529-1540.
51. Chang BS, Patro SY. Freeze-drying process development for protein pharma- 83. Wolfe J, Bryant G. Freezing, drying, and/or vitrification of mem-
ceuticals. In: Lyophilization of Biopharmaceuticals. 2. 2004:113. braneesoluteewater systems. Cryobiology. 1999;39(2):103-129.
52. Chen H, Khemtong C, Yang X, Chang X, Gao J. Nanonization strategies for 84. Salazar J, Ghanem A, Müller RH, Mo €schwitzer JP. Nanocrystals: comparison of
poorly water-soluble drugs. Drug Discov Today. 2011;16(7e8):354-360. the size reduction effectiveness of a novel combinative method with con-
53. Müller RH, Peters K. Nanosuspensions for the formulation of poorly soluble ventional top-down approaches. Eur J Pharm Biopharm. 2012;81(1):82-90.
drugs: I. Preparation by a size-reduction technique. Int J Pharm. 1998;160(2): 85. Teagarden DL, Baker DS. Practical aspects of lyophilization using non-aqueous
229-237. co-solvent systems. Eur J Pharm Sci. 2002;15(2):115-133.
M. Mohammady et al. / Journal of Pharmaceutical Sciences 109 (2020) 3235-3247 3247

86. Mason TG, Wilking JN, Meleson K, Chang CB, Graves SM. Nanoemulsions: 117. Eddleston MD, Patel B, Day GM, Jones W. Cocrystallization by freeze-drying:
formation, structure, and physical properties. J Phys Condens Matter. preparation of novel multicomponent crystal forms. Cryst Growth Des.
2006;18(41):R635. 2013;13(10):4599-4606.
87. Aboofazeli R. Nanometric-scaled emulsions (nanoemulsions). Iran J Pharm Res. 118. Grossjohann C, et al, Serrano DR, Paluch KJ. Polymorphism in sulfadimidine/4-
2010;9(4):325. aminosalicylic acid cocrystals: solid-state characterization and physico-
88. Taylor KM, Aulton ME. Aultons Pharmaceutics: The Design and Manufacture of chemical properties. J Pharm Sci. 2015;104(4):1385-1398.
Medicines. Elsevier; 2013. 119. Zhang D, Tan T, Gao L, Zhao W, Wang P. Preparation of azithromycin nano-
89. McClements DJ. Nanoemulsions versus microemulsions: terminology, differ- suspensions by high pressure homogenization and its physicochemical
ences, and similarities. Soft Matter. 2012;8(6):1719-1729. characteristics studies. Drug Dev Ind Pharm. 2007;33(5):569-575.
90. Zhang Y, Shang Z, Gao C, et al. Nanoemulsion for solubilization, stabilization, 120. Liu D, Xu H, Tian B, et al. Fabrication of carvedilol nanosuspensions through
and in vitro release of pterostilbene for oral delivery. AAPS PharmSciTech. the anti-solvent precipitationeultrasonication method for the improvement
2014;15(4):1000-1008. of dissolution rate and oral bioavailability. AAPS Pharm Sci Tech. 2012;13(1):
91. Capek I. Degradation of kinetically-stable o/w emulsions. Adv Colloid Interface 295-304.
Sci. 2004;107(2-3):125-155. 121. Van Eerdenbrugh B, Froyen L, Martens J, et al. Characterization of physico-
92. Gupta S, Chavhan S, Sawant KK. Self-nanoemulsifying drug delivery system chemical properties and pharmaceutical performance of sucrose co-
for adefovir dipivoxil: design, characterization, in vitro and ex vivo evaluation. freezeedried solid nanoparticulate powders of the anti-HIV agent loviride
Colloid Surface Physicochem Eng Aspect. 2011;392(1):145-155. prepared by media milling. Int J Pharm. 2007;338(1e2):198-206.
93. Komatsu H, Saito H, Okada S, Tanaka M, Egashira M, Handa T. Effects of the 122. Ibrahim AH, Rosqvist E, Smått J-H, et al. Formulation and optimization of
acyl chain composition of phosphatidylcholines on the stability of freeze- lyophilized nanosuspension tablets to improve the physicochemical properties
dried small liposomes in the presence of maltose. Chem Phys Lipids. and provide immediate release of silymarin. Int J Pharm. 2019;563:217-227.
2001;113(1e2):29-39. 123. Kanthamneni N, Valiveti S, Patel M, Xia H, Tseng YC. Enhanced bioavailability
94. McClements D, Decker E, Weiss J. Emulsion-based delivery systems for lipo- of danazol nanosuspensions by wet milling and high-pressure homogeniza-
philic bioactive components. J Food Sci. 2007;72(8):R109-R124. tion. Int J Pharm Investig. 2016;6(4):218.
95. Hedoux A, Paccou L, Achir S, Guinet Y. Mechanism of protein stabilization by 124. Oktay AN, Karakucuk A, Ilbasmis-Tamer S, Celebi N. Dermal flurbiprofen
trehalose during freeze-drying analyzed by in situ micro-Raman spectros- nanosuspensions: Optimization with design of experiment approach and in
copy. J Pharm Sci. 2013;102(8):2484-2494. vitro evaluation. Eur J Pharm Sci. 2018;122:254-263.
96. do Vale Morais AR, do Nascimento Alencar E,  Júnior FHX, et al. Freeze-drying 125. Lyu J, Zhang H, Wang L, Gao J. Preparation and Dissolution of Loratadine
of emulsified systems: a review. Int J Pharm. 2016;503(1e2):102-114. Nanosuspension Lyophilized Powder. China Pharmacist. 2018;21(5):809-813.
97. Li F, Wang T, He HB, Tang X. The properties of bufadienolides-loaded nano- 126. He J, Han Y, Xu G, et al. Preparation and evaluation of celecoxib nanosuspensions
emulsion and submicro-emulsion during lyophilization. Int J Pharm. for bioavailability enhancement. RSC Adv. 2017;7(22):13053-13064.
2008;349(1e2):291-299. 127. Rao YM, Kumar MP, Apte S. Formulation of nanosuspensions of albendazole
98. Wei W, Mo C, Guohua C. Issues in freeze drying of aqueous solutions. Chin J for oral administration. Curr Nanosci. 2008;4(1):53-58.
Chem Eng. 2012;20(3):551-559. 128. Liversidge GG, Cundy KC. Particle size reduction for improvement of oral
99. Couvreur P, Barratt G, Fattal E, Vauthier C. Nanocapsule technology: a review. bioavailability of hydrophobic drugs: I. Absolute oral bioavailability of
Crit Rev Ther Drug Carrier Syst. 2002;19(2). nanocrystalline danazol in beagle dogs. Int J Pharm. 1995;125(1):91-97.
100. Anton N, Benoit J-P, Saulnier P. Design and production of nanoparticles 129. Ain-Ai A, Gupta PK. Effect of arginine hydrochloride and hydroxypropyl cellulose
formulated from nano-emulsion templatesda review. J Control Release. as stabilizers on the physical stability of high drug loading nanosuspensions of a
2008;128(3):185-199. poorly soluble compound. Int J Pharm. 2008;351(1e2):282-288.
101. Letchford K, Burt H. A review of the formation and classification of amphi- 130. Lou H, Zhang X, Gao L, et al. In vitro and in vivo antitumor activity of oridonin
philic block copolymer nanoparticulate structures: micelles, nanospheres, nanosuspension. Int J Pharm. 2009;379(1):181-186.
nanocapsules and polymersomes. Eur J Pharm Biopharm. 2007;65(3):259-269. 131. Teeranachaideekul V, Junyaprasert VB, Souto EB, Müller RH. Development of
102. Nakagawa K, Surassmo S, Min SG, Choi MJ. Dispersibility of freeze-dried poly ascorbyl palmitate nanocrystals applying the nanosuspension technology. Int
(epsilon-caprolactone) nanocapsules stabilized by gelatin and the effect of J Pharm. 2008;354(1e2):227-234.
freezing. J Food Eng. 2011;102(2):177-188. 132. de Waard H, De Beer T, Hinrichs WL, Vervaet C, Remon JP, Frijlink HW.
103. Bejrapha P, Min SG, Surassmo S, Choi MJ. Physicothermal properties of freeze- Controlled crystallization of the lipophilic drug fenofibrate during freeze-
dried fish oil nanocapsules frozen under different conditions. Dry Technol. drying: Elucidation of the mechanism by in-line Raman spectroscopy. AAPS J.
2010;28(4):481-489. 2010;12(4):569-575.
104. Searles JA, Carpenter JF, Randolph TW. Annealing to optimize the primary 133. Ma YQ, Li G, Xu JH, et al. Combination of submicroemulsion and phospholipid
drying rate, reduce freezing-induced drying rate heterogeneity, and deter- complex for novel delivery of ursodeoxycholic acid. Pharm Dev Technol.
mine Tg' in pharmaceutical lyophilization. J Pharm Sci. 2001;90(7):872-887. 2014;19(3):363-372.
105. Abdelwahed W, Degobert G, Fessi H. Freeze-drying of nanocapsules: impact 134. Gol D, Thakkar S, Misra M. Nanocrystal-based drug delivery system of ris-
of annealing on the drying process. Int J Pharm. 2006;324(1):74-82. peridone: lyophilization and characterization. Drug Dev Ind Pharm.
106. Bexiga NM, Bloise A, Alencar AM, Stephano MA. Freeze-drying of ovalbumin- 2018;44(9):1458-1466.
loaded carboxymethyl chitosan nanocapsules: impact of freezing and 135. Khayata N, Abdelwahed W, Chehna MF, Charcosset C, Fessi H. Preparation of
annealing procedures on physicochemical properties of the formulation vitamin E loaded nanocapsules by the nanoprecipitation method: From lab-
during dried storage. Dry Technol. 2018;36(4):400-417. oratory scale to large scale using a membrane contactor. Int J Pharm.
107. Kamiya S, Kurita T, Miyagishima A, Itai S, Arakawa M. Physical properties of 2012;423(2):419-427.
griseofulvin-lipid nanoparticles in suspension and their novel interaction 136. Torrecilla D, Lozano MV, Lallana E, et al. Anti-tumor efficacy of chitosan-g-
mechanism with saccharide during freeze-drying. Eur J Pharm Biopharm. poly (ethylene glycol) nanocapsules containing docetaxel: anti-TMEFF-2
2010;74(3):461-466. functionalized nanocapsules vs. non-functionalized nanocapsules. Eur J Pharm
 
108. Curi c A, Keller BL, Reul R, Mo €schwitzer J, Fricker G. Development and Biopharm. 2013;83(3):330-337.
lyophilization of itraconazole loaded poly (butyl cyanoacrylate) nanospheres 137. Surassmo S, Min SG, Bejrapha P, Choi MJ. Effects of surfactants on the physical
as a drug delivery system. Eur J Pharm Sci. 2015;78:121-131. properties of capsicum oleoresin-loaded nanocapsules formulated through
109. de Chasteigner S, Cave  G, Fessi H, Devissaguet JP, Puisieux F. Freeze-drying of the emulsionediffusion method. Food Res Int. 2010;43(1):8-17.
itraconazole-loaded nanosphere suspensions: a feasibility study. Drug Dev 138. Lu B, Xiong S-B, Yang H, Yin X-D, Zhao RB. Mitoxantrone-loaded BSA nano-
Res. 1996;38(2):116-124. spheres and chitosan nanospheres for local injection against breast cancer
110. He W, Lu Y, Qi J, Chen L, Hu F, Wu W. Nanoemulsion-templated shell-cross- and its lymph node metastases: I: Formulation and in vitro characterization.
linked nanocapsules as drug delivery systems. Int J Pharm. 2013;445(1e2):69- Int J Pharm. 2006;307(2):168-174.
78. 139. Pfeifer BA, Burdick JA, Langer R. Formulation and surface modification of poly
111. Lozano M, Esteban H, Brea J, Loza M, Torres D, Alonso M. Intracellular delivery (ester-anhydride) micro-and nanospheres. Biomaterials. 2005;26(2):117-124.
of docetaxel using freeze-dried polysaccharide nanocapsules. J Microencapsul. 140. Donini C, Robinson D, Colombo P, Giordano F, Peppas N. Preparation of poly
2013;30(2):181-188. (methacrylic acid-g-poly (ethylene glycol)) nanospheres from methacrylic
112. Oyarzun-Ampuero FA, Rivera-Rodríguez GR, Alonso MJ, Torres D. Hyaluronan monomers for pharmaceutical applications. Int J Pharm. 2002;245(1e2):83-
nanocapsules as a new vehicle for intracellular drug delivery. Eur J Pharm Sci. 91.
2013;49(4):483-490. 141. Wagh P, Mujumdar A, Naik JB. Preparation and characterization of ketorolac
113. Allu S, Bolla G, Tothadi S, Nangia A. Supramolecular synthons in bumetanide tromethamine-loaded ethyl cellulose micro-/nanospheres using different
cocrystals and ternary products. Cryst Growth Des. 2017;17(8):4225-4236. techniques. Particul Sci Technol. 2019;37(3):347-357.
114. Desiraju GR. Supramolecular synthons in crystal engineeringda new organic 142. ElShagea HN, ElKasabgy NA, Fahmy RH, Basalious EB. Freeze-Dried Self-
synthesis. Angew Chem. 1995;34(21):2311-2327. Nanoemulsifying Self-Nanosuspension (SNESNS): a New Approach for the
115. Zhao L, Raval V, Briggs NE, et al. From discovery to scale-up: a-lipoic acid: Preparation of a Highly Drug-Loaded Dosage Form. AAPS Pharm Sci Tech.
nicotinamide co-crystals in a continuous oscillatory baffled crystalliser. 2019;20(7):258.
CrystEngComm. 2014;16(26):5769-5780. 143. Wang L, Zhao X, Zu Y, et al. Enhanced dissolution rate and oral bioavailability
116. Rodrigues M, Baptista B, Lopes JA, Sarraguça MC. Pharmaceutical cocrystalli- of ginkgo biloba extract by preparing nanoparticles via emulsion solvent
zation techniques. Advances and challenges. Int J Pharm. 2018;547(1e2):404- evaporation combined with freeze drying (ESE-FR). Rsc Adv. 2016;6(81):
420. 77346-77357.

You might also like