You are on page 1of 15

International Journal of Pharmaceutics 586 (2020) 119555

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


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

Review

Formulation aspects of intravenous nanosuspensions T


Dipeekakumari Patel, Sandeep S. Zode, Arvind K. Bansal

National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India

ARTICLE INFO ABSTRACT

Keywords: Intravenous (IV) route is preferred for rapid onset of action, avoiding first pass metabolism and achieving site
Intravenous delivery specific delivery. Development of IV formulations for poorly water soluble drugs poses significant challenges.
Nanocrystalline suspensions Formulation approaches like salt formation, co-solvents, surfactants and inclusion complexation using cyclo-
Scale-up dextrins are used for solubilisation. However, these approaches are not applicable universally and have lim-
Tissue targeting
itations in extent of solubilisation, hypersensitivity, toxicity and application to only specific type of molecules. IV
Regulatory
nanosuspension have been attracting attention as a viable strategy for development of IV formulations of poorly
water-soluble drugs. Nanosuspension consists of nanocrystals of poorly water soluble drug suspended in aqueous
media and stabilized using minimal concentration of stabilizers. Recent years have witnessed their potential in
formulations for toxicological studies and clinical trials. However various challenges are associated with the
translational development of IV nanosuspensions. Therefore, the objective of the current review is to provide a
holistic view of formulation development and desired properties of IV nanosuspensions. It will also focus on
advancements in characterization tools, manufacturing techniques and post-production processing. Challenges
associated with translational development and regulatory aspects of IV nanosuspension will be addressed.
Additionally, their role in preclinical evaluation and special applications like targeting will also be discussed
with the help of case studies. The applications of IV nanosuspensions shall expand as their applications move
from preclinical phase to commercialization.

1. Introduction drugs, co-solvents, surfactants and inclusion complexes like cyclodex-


trins (Wong et al., 2008). Use of salt form of drug is only applicable to
New chemical entities (NCE) coming into pipeline are poorly water ionizable drugs and there is risk of in-vivo precipitation due to change in
soluble due to the emerging role of computational and combinatorial pH. (Bracq et al., 2008). Hemotoxicity is associated with use of some
chemistry (Kipp, 2004). It has been observed that 90% of these new co-solvents such as ethanol and polyethylene glycol while higher
chemical entities belong to Biopharmaceutics Classification System amounts of surfactants (like Cremophor) can lead to hypersensitivity
(BCS) class II (70%) or class IV (20%). Classification of BCS class II as reactions like pseudo-anaphylactoid characterized by pruritus, er-
per the developability classification system (DCS) follows two sub- ythema, rash or generalized urticaria (ten Tije et al., 2003). Cyclodex-
classes: dissolution rate limited class IIa, termed as ‘brick-dust’ mole- trins used in higher concentrations increases the viscosity of solutions
cules, and solubility limited class IIb, the so-called ‘grease-ball’ mole- which is unfavourable in case of IV formulation and can cause ne-
cules (Butler and Dressman, 2010). Brick-dust molecules are phrotoxicity on prolonged use. They are also not preferred when the
characterized by high melting point of > 200 °C and moderate lipo- dose of drug is high, as the molar ratio of the cyclodextrins and the drug
philicity (log P < 2). They display poor solubility in water as well as is generally 1:1 or 2:1 (Yalkowsky, 1999). IV nanosuspension can be a
lipids (eg. itraconazole). Grease-ball molecules have log P of > 3 and viable formulation option for brick-dust molecules having high dose
low melting points eg. danazol (log P = 4.53), tamoxifen (log and grease-ball molecules having high melting point.
P = 5.26) (Fagerberg et al., 2010) and show solubility in at least some Nanosuspension consists of nanocrystals of poorly water soluble
lipids (Bergström et al., 2016). For grease-ball molecules, a lipidic drug suspended in aqueous media and stabilized using low concentra-
system or an intravenous emulsion can be adopted, provided the drug tion of stabilizers (Patravale et al., 2004). They have particle size in the
has low melting point (Wong et al., 2008). nano scale range (below 1 µm) and thus have potential advantage over
Formulation strategies to solubilize the brick-dust molecules for an coarse suspension. The small particle size shows significantly rapid
intravenous (IV) formulation include pH adjustment, using salt form of dissolution. Clinically the major benefits of a nanosuspension


Corresponding author.
E-mail address: akbansal@niper.ac.in (A.K. Bansal).

https://doi.org/10.1016/j.ijpharm.2020.119555
Received 14 March 2020; Received in revised form 23 May 2020; Accepted 14 June 2020
Available online 17 June 2020
0378-5173/ © 2020 Elsevier B.V. All rights reserved.
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

Fig. 1. Various technologies for generations of nanocrystals (Sheokand et al., 2018).

formulation include (1) high drug loading that facilitates low volume of be categorized into bottom-up, top-down and combination approaches.
injection, (2) low toxicity due to absence of solubilizing agents, (3) Bottom-up technique involves self-assembling of particles to form na-
possible alteration of the pharmacokinetics (PK) of the drug with noparticles and includes dissolving the drug in an organic solvent fol-
modified drug release formulations and (4) targeted delivery of drug by lowed by adding an antisolvent to precipitate the drug in presence of a
either active or passive targeting. IV nanosuspension have been at- stabilizer. Organic solvents and antisolvents are miscible with each
tracting lot of attention because of aforementioned potential benefits other. Commonly used solvents include ethanol, dimethylsulfoxide,
(Pace et al., 1999; Rabinow, 2004). Successful formulations have been methanol, dichloromethane and acetone, while water is the most
reported for antineoplastic agents (Merisko-Liversidge et al., 1996), commonly used antisolvent. Bottom-up approaches include super-
anaesthetic agents (Boedeker et al., 1994), antifungals (Rabinow et al., critical fluid processes, sonocrystallization, liquid jet crystallization,
2007) and agents for malignant hyperthermia (Karan et al., 1996). emulsion solvent evaporation and spray drying.(Ahire et al., 2018; Suri
IV nanosuspension are a popular formulation approach during de- et al., 2016) More recently, a novel bottom-up technique to produce
velopment of drug molecules especially when the safety of the NCE nanocrystals was developed by controlled crystallization using freeze-
needs to be assessed in experimental animals at multi-fold times of its drying.(De Waard et al., 2008).
intended human dose. It is possible to eliminate the toxic adverse ef- In top-down technique, larger particles are reduced to nanoparticles
fects of co-solvents and administer significantly larger doses of drug by size reduction techniques. This mainly includes the wet media mil-
(Kesisoglou and Mitra, 2012). Recent years have witnessed their ap- ling (WMM) technique, high pressure homogenization (HPH) and mi-
plications in toxicological and clinical formulations (Frank and Boeck, crofluidization technique. In WMM, the coarse drug particles are
2016). Therefore, the drug molecules once abandoned due to solubility charged in a dispersion medium containing milling beads and stabilizer
and toxicity issues can now be explored for product development. Al- (s). Particle size reduction is mainly due to mechanical attrition(Gao
though literature is available on nanosuspension in general, very few et al., 2008a). Contamination of the nanocrystalline suspension can be
reports are available on IV nanocrystalline suspension which can be of due to abrasion of milling glass beads or zirconium beads or parts of
two types i.e. ‘ready to use’ and ‘dry powder nanosuspension for re- milling chamber. This contamination limits the use of WMM method for
constitution’. This review will focus on formulation development, de- an IV product. However, this challenge was overcome by introduction
sired properties, analytical aspects and challenges associated with of highly crosslinked polystyrene beads as milling media (Kesisoglou
translational development of IV nanocrystalline suspension. Ad- et al., 2007). This media undergoes elastic deformation, thus reducing
ditionally, their role in preclinical evaluation, special applications like formation of cracks and abrasions. The commercial technology Nano-
targeting and regulatory aspects will be addressed. Crystal® uses PolyMill media which comprises of polystyrene beads.
This leads to nanosuspension with high product purity which can be
used parenterally. Invega Sustenna, a nanocrystal based product, was
2. Manufacturing techniques generated using WMM based NanoCrystal® technology.
HPH is an alternative technique for the production of nanocrystals
Nanosuspension can be prepared using three approaches. These can

2
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

by top-down approach. In this technique, drug suspension passes at a free radical formation by ionizing radiation. Therefore, it is re-
high speed through the narrow orifice of the homogenizer. Particle size commended to down process the formulation to a dry powder and then
reduction is brought about by cavitation, high shear forces and the sterilize it using gamma radiation (Reid, 1995). A radiation dose of
collision of the particles against each other (Müller et al., 2011). An- 25 kGy is recommended for sterilization of nanosuspension (Shegokar
other technique is microfluidization technique where two fluid streams and Singh, 2012). Sterilization method best suited for the formulation
collide at very high pressure resulting in generation of nanocrystals. It is must be chosen on the basis of formulation components.
based on the principle of jet-stream homogenization (Shegokar and
Müller, 2010). Combination techniques simply uses both top-down and 3.1.3. Redispersibility and reconstitution time
bottom-up technologies to generate nanosuspension. Fig. 1 outlines the Sterile powders for reconstitution as nanosuspension can be formed
various methods that are used in the preparation of nanosuspension after downstream processing of the nanosuspension formulation either
along with their principles, advantages and disadvantages. For detailed by lyophilization or other technique. These powders are supplied with a
explanation of manufacturing techniques, readers can refer the reviews suitable sterile diluent (usually water for injection) for reconstitution as
by Patravale et. al. (Patravale et al., 2004), Lakshmi et. al. (Lakshmi and nanosuspension. It is, therefore, essential that the nanocrystals redis-
Kumar, 2010), Salazar et. al. (Salazar et al., 2014). perse easily with gentle agitation and the nanocrystals maintain their
integrity after reconstitution. USP mentions specific tests of uniformity
3. Development of IV nanosuspensions: of dosage units and water content for sterile powders for suspension
which can be taken into consideration for nanosuspension as well.
3.1. Desired formulation properties
3.1.4. Osmolality
3.1.1. Particle size The determination of the osmolar concentration of plasma is called
Particle size of the nanocrystals is a crucial factor for its safety and osmolality and it is proportional to the number of particles per kilogram
in vivo fate. Drug nanocrystals will come across the venules (lumen of solvent. It is usually expressed as mOsmol/kg. As per USP, the os-
diameter of ~ 20 µm) and capillaries (lumen diameter of ~ 6 µm), molality of blood should range between 285 and 310 mOsmol/kg.
following IV administration (Wong et al., 2008). Particles > 7 µm and Therefore, the average physiological osmolality is approximately 297.5
larger agglomerates can cause pulmonary embolism and may risk the mOsmol/kg (Roethlisberger et al., 2017). It has been reported that
patient safety. Therefore, the particle size in a nanocrystalline suspen- hypertonic solutions with an osmolality > 600 mOsmol/kg can cause
sion should be below 5 µm. It was reported that etoposide-bovine serum shrivelling up of the erythrocytes and cause pain. While hypotonic in-
albumin nanosuspension when injected IV (particle sizes of jection solutions with an osmolality of < 150 mOsmol/kg may cause
100–300 nm) was taken up by the tumor cells by means of enhanced haemolysis and pain at the injection site. The limits for osmolality have
permeation and retention effect (Xiong et al., 2008). Size of the crys- to be interpreted with caution because there are several factors like
talline particles can influence the pharmacokinetic behaviour of the selection of vein for injection, injection volume and duration that can
formulation. For example, animal studies conducted with itraconazole cause irritation (Roethlisberger et al., 2017). There are products with
nanosuspension indicated size dependent pharmacokinetic perfor- significant hyperosmolality and the tolerance values differs with infu-
mance which was most distinct for larger crystals (i.e., sion and small volume injection. Therefore, for drug products intended
those ≥ 340 nm). Size of approximately 100–200 nm is preferred for for intravenous use, the recommended upper osmolality limit should be
rapid dissolution. The pharmacokinetics of such nanosuspension is ex- controlled generally under 1000 mOsmol/kg (Wang, 2015).
pected to be similar to that of an IV solution. When prolonged dis-
solution is required, the mean particle diameter must be in the rela- 3.1.5. pH
tively greater nanometer range, e.g. 800–1000 nm. This prolonged A formulation with an extreme pH can possibly cause inflammatory
effect helps in targeting of nanosuspension to the monocyte phagocytic reactions like vascular irritation or pain. Therefore, a target pH range of
system (MPS) cells by avoiding complete dissolution of the particles 3.5–9.0 is recommended for small volume injections to avoid such
before they reach the macrophages (Müller et al., 2001). complications (Roethlisberger et al., 2017). Buffering agents are used to
adjust the pH will be discussed in latter section. The physiological local
3.1.2. Sterility reaction cascade on IV injection depends on a number of factors like
Sterility is one of the most important criteria for any IV formulation. injection volume, injection rate, blood flow, viscosity, cosolvents and
Sterilization of injectables is achieved either through aseptic proces- active agents. Therefore, a direct correlation between pain or irritation
sing, terminal sterilization using autoclaving or gamma radiation (for and pH cannot be made unless all such factors are kept constant. In case
dry powders). However, nanosuspension offer a unique challenge as of a marginal pH, a slow infusion rate (for example, 5-minute push
these methods may deleteriously affect the formulation and its stability. instead of a quick bolus injection) might help to minimize the risk of
For example, Shegokar et al. investigated the effect of radiation and vascular irritation and vein damage. Better tolerance in this case can be
moist heat sterilization on the lyophilized nanosuspension. They con- attributed to the slow infusion time and to the increased dilution of
cluded that the mean particle size increased with the escalation of ra- drug by the flow of blood at the injection site (Roethlisberger et al.,
diation dose and with moist heat sterilization (Shegokar and Singh, 2017). IV drug products with a wide range of pH values (2.55–11.15)
2012). Therefore, we need to devise product-specific techniques for are available in the market as they overcome the drug solubility and
sterilization. One way is to sterilize the API and the excipients sepa- stability issues. This shows that one can optimize the pH of the IV na-
rately and mix them under aseptic conditions. The raw material drug nosuspension in the above mentioned range based on the formulation
can be obtained as a sterile grade API or it can be sterilized by gamma development requirement. However, for drugs having pH dependent
radiation (Reid, 1995) or by dry heat (Moller and Jensen, 1970) if the solubility, pH should be kept in a narrow range to prevent Ostwald-
drug is stable to either of the methods. An aqueous solution containing ripening.
excipients can be separately sterilized by conventional means and then
mixed with the API under aseptic condition. Another strategy can be 3.1.6. Endotoxins and pyrogenicity
sterile filtration of aqueous nanosuspension prepared using sterile API. Endotoxins are lipopolysaccharides found in the outer membrane of
An example of sterile filtration for nanosuspension is the NanoCrsytalTM gram negative bacteria and their presence even in nanogram quantities
for X-ray contrast agent iodipamide. 0.2 µm Supor® 200 DCF™ filter was can elicit immunological response. The endotoxin limit for parenteral
used and 100% of Pseudomonas diminuta were retained through it formulation is defined on the basis of dose and is given by the equation
(Zheng and Bosch, 1997). Radiation sterilization is unacceptable due to K/M, where K is the threshold human pyrogenic dose of endotoxin per

3
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

kg of body weight, and M is the maximum recommended human dose Table 1


per kg of body weight per hour. The endotoxin limit for API used in the Stabilizers used in IV formulations and their IID limits.
formulation must be known at the start of formulation. For parenteral Sr. No. Stabilizer IID limit (Maximum potency
drugs, the endotoxin limit is specified in individual monographs and per unit dose)
expressed in units like EU/ml or EU/unit. In absence of any active
1. Poloxamer 188 0.6% w/v
procedure of subsequently removing the endotoxins, it is important to
2. Polyethylene glycol 400 75.58% w/v
limit the total endotoxins by controlling the specifications of input 3. Polysorbate 20 4.8% w/v
materials in the formulation. An in-house specification can be included, 4. Polysorbate 80 69.33% w/v
to meet the general criteria of injectables. USP Chapter 85 describes the 5. Povidone K12 10% w/v
Bacterial Endotoxins Test based on the use of Limulus Amoebocyte 6. Soybean lecithin 21.3% w/v
7. D-α-Tocopheryl polyethylene glycol 225 mg/L
Lysate (LAL) reagent to detect bacterial endotoxins in the parenteral
1000 succinate (TPGS)
formulation (Chapter85, 2007). This chapter includes the photometric 8. Albumin 2% w/v
techniques (chromogenic and turbidimetric tests) and gel-clot techni-
ques using LAL reagent. Gel-clot technique is more commonly used and
the reaction endpoint is determined by comparison with a reference stabilizers to the particle surface (Verma et al., 2009a). Stabilizer se-
standard. Precautions should be taken with sample preparations and lection can also be done according to their surface tension. The study
dilutions of the test sample and standard sample with LAL reagent. can be performed by contact angle measurement. The stabilizer with
Pyrogenicity testing is an alternative to bacterial endotoxin test. USP lower surface tension and low contact angle acts as a better stabilizer
pyrogen test provides information about requirements for the absence (Chen et al., 2004). The amount and type of stabilizer used in the na-
of pyrogens in a parenteral formulation. The test involves measurement nosuspension formulation has a prominent effect on the physical sta-
of rise in temperature of rabbits following IV administration of drug bility and in-vivo performance of the formulation. Regardless of the
formulation and is limited for products that can be tolerated by test method (bottom-up or top-down or combination) used for the pre-
rabbits in a dose that do not exceed 10 mL per kg injected IV within a paration, the selection of the right type of stabilizer with its optimal
period of not > 10 min. Before the initiation of the test, care should be concentration are essential for stabilizing the smaller size particles. This
taken that all of the excipients used must meet USP limits on endotoxins would help to maintain the shelf life stability of the final product (Van
and all the glasswares are depyrogenated using a validated method. Eerdenbrugh et al., 2009; Wu et al., 2011). However, researchers have
Test nanosuspension formulation must be injected IV into three rabbits, demonstrated the preparation of nanoparticles without using any sta-
the formulation passes the test if none of the animals demonstrate an bilizer. This preparation requires rapid and powerful mixing devices
individual temperature rise of 0.5 °C or more to its corresponding (Wang et al., 2007; Xu et al., 2012).
control temperature for 3 h. A retest procedure has to be followed if the
test fails. The pyrogen test can be used for product release.
3.2.2. Organic solvents
Bottom up process and combination approaches used for generation
3.2. Formulation components
of nanosuspension utilize organic solvents. Organic solvent like
ethanol, isopropyl alcohol, methanol, acetone and N-Methylpyrrolidone
Developing an IV nanosuspension may be challenging as the
can be used for preparing the drug solution (Dong et al., 2009). How-
number of excipients approved for parenteral route are limited in
ever, residual organic solvents may raise toxicity concerns and increase
numbers. The formulation may comprise of stabilizers, buffering
regulatory burden. According to ICH guidelines, class lll solvents can be
agents, tonicity adjusting agents and organic solvents.
used in the preparation of nanocrystals as these solvents are regarded as
less toxic. Class II solvents can also be used if their residual solvent
3.2.1. Stabilizers
remains within the permissible limits. Class I are considered toxic and
Stabilizers are used to wet the drug particles and to prevent
cannot be used in IV formulations. Table 2 consists of the ICH class II
Ostwald’s ripening (Müller et al., 1998). Absence of the stabilizers can
and III organic solvents along with their permissible daily exposure
induce formation of aggregates, due to high surface energy of drug
(PDE) and concentration limits.
nanocrystals. They should be selected based on their interaction with
active pharmaceutical ingredient (API) and their stabilization potential.
Stabilizers can be either ionic surfactants, known to exhibit electrostatic 3.2.3. Buffers
repulsion or polymers which provide steric hindrance and thereby A physiological pH (~7.4) should be the target for IV formulations
prevent particle aggregation. A combination of surfactants and poly- as extreme pH (< pH 3 or > pH 10.5) may cause irritation upon ad-
mers can be used to provide better stabilization. Stabilizers should be ministration due to local cell damage and necrosis (Roethlisberger
selected on the basis of FDA’s Inactive Ingredient Database (IID) for IV et al., 2017). pH of the formulation needs to be controlled to avoid drug
route. Some of the stabilizers that have been explored for IV formula- degradation during processing, storage and reconstitution therefore, it
tions include Pluronic F-68, Polysorbate 20, Lecithin, Polysorbate 80, becomes important to evaluate buffering agents. Selection of the buf-
Pluronic F-108 and vitamin E polyethylene glycol succinate (TPGS) (Li fering agent depends on the pH stability of the API. However, the
et al., 2011). List of FDA approved stabilizers along with their IID limits
for IV route are given in table 1. The ratio of drug and stabilizer may Table 2
vary from 1:20 to 20:1 and must be investigated depending on the ICH class solvents with their PDE concentration limits.
physicochemical properties of API (George and Ghosh, 2013). Further, Solvent Class of PDE (mg/ Concentration limit
surfactant concentration can be decided based on its critical micelle solvent day) (ppm)
concentration (CMC). Care should be taken not to cross the CMC of the
Acetonitrile 2 4.1 410
surfactants, as above CMC micelle formation takes place and surface 1,4- Dioxane 2 3.8 380
adsorption on particles is reduced. Moreover, this leads to increased Methanol 2 30 3000
particle size as micelle formation increases the solubility of API thereby N-Methylpyrrolidone 2 5.3 530
enhancing particle growth by Ostwald ripening (Sinha et al., 2013). Acetone 3 50 5000
Dimethyl sulfoxide 3 50 5000
Toxicity of the surfactants above CMC is also a concern for IV for-
Ethanol 3 50 5000
mulations. Isopropyl alcohol 3 50 5000
Atomic force microscopy (AFM) can be used to study the affinity of

4
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

Table 3 lyophilization, particle size of the nanocrystals may increase. In a study,


List of buffering agents used in IV formulations. the authors reported that the particle size was not maintained after
Buffering agents pKa pH range lyophilization. However, the final size obtained was still below 5 µm
which suggested that it could be administered intravenously (Shegokar
Maleic acid 1.9, 6.2 2–3 and Singh, 2012). Similar results of particle size increments were ob-
Tartaric acid 2.9, 4.2 2.5–4
tained when doxorubicin loaded chitosan nanoparticles were lyophi-
Lactic acid 3.8 3–4.5
Citric acid 3.1, 4.8, 6.4 3–7
lized with mannitol and sucrose as cryoprotectant (Dave et al., 2012).
Acetic acid 4.75 4–6 In one of the studies reported, it was observed that after reconstituting
Sodium bicarbonate 6.3, 10.3 4–9 the lyophilized product, the mean size of Ascorbyl palmitate nanosus-
Sodium phosphate 2.2, 7.2, 12.4 6–8 pensions without cryoprotectant (particle size~540 nm) was sig-
Sodium hydroxide 13.8 12
nificantly higher compared with the system containing trehalose as
Glycine 2.34, 9.6 2.2–3.6; 8.8–10.6
Histidine 1.82, 9.17 5.5–7.4 cryoprotectant (1–10% w/w) (particle size of ~360 nm)
(Teeranachaideekul et al., 2008). Therefore, it can be concluded that
the type and amount of cryoprotectant added to the formulation has a
interaction of the buffer with the solvent system and the drug should be pronounced effect on particle size of the freeze-dried product (Shegokar
examined as they are likely to affect surface charge or may interact with and Singh, 2011).
the surfactants/ drug, thus changing their properties. The following pH shifts in the formulation during lyophilization may adversely
table 3 comprises of the FDA approved buffering agents that can be affect the product thereby making it unsuitable for intravenous ad-
commonly used in IV formulations. ministration. Buffering species present in the formulation may crystal-
lize during the process of lyophilization leading to a drastic shift in pH,
3.2.4. Tonicity adjusting agents resulting in degradation of the active component. Sodium and po-
Isotonicity is another key requirement, particularly if the product is tassium phosphate salts crystallize during cooling and in frozen solution
intended for large volume infusion, as hypotonic solutions cause hae- which leads to a decrease in pH of about 4 units and therefore they are
molysis. Hypertonic solutions may be tolerated if administered slowly not used in lyophilization (Gómez et al., 2001). Shalaev et al. studied
in small volumes (Shi et al., 2009). Non-ionic excipients like mannitol succinate, citrate and tartrate buffers for their crystallization behaviour
and dextrose have been effectively applied without affecting the PSD and its effect on pH of the formulation. It was found that citrate buffer
for adjusting the tonicity (Jacobs et al., 2000; Sigfridsson et al., 2007). was better in comparison to succinate and tartrate as it remained
Moreover, they provide additional advantage of acting as matrix amorphous, with pH shift being minimal, while the others crystallized
forming agent in the downstream processing of the nanosuspension during lyophilization (Shalaev et al., 2002). It is advisable to place the
through spray-drying or freeze drying (discussed in detail in subsequent buffering agent in the reconstitution fluid if it interacts with the active
section). Salts are usually avoided to adjust the tonicity of the nano- component or hinders the lyophilization process.
crystalline suspension as they may destabilize the formulation by al-
tering the electric charge of the nanoparticles. 4.2. Spray drying

Spray drying is routinely used for the conversion of liquid for-


4. Dry powder nanosuspension for reconstitution mulation into dry solid powder. In this process, nanosuspension is
atomised to fine droplets in a preheated chamber which are then eva-
Post-production processing, also known as downstream processing, porated to form dry powder. Heat and mass transfer occur between air
of nanosuspension is crucial when the drug is extremely susceptible to and the particles which might lead to slight stickiness depending upon
chemical degradation. It may also be essential when the stabilizer the formulation composition. This stickiness or aggregation can be
cannot effectively stabilize the formulation for a longer period of time circumvented by adding adsorbents such as lactose, mannitol, mal-
and the particle size increases as a result of Ostwald ripening. Other todextrose, etc.
challenges associated with nanosuspension are physical instabilities The nature of adsorbents with their concentration and spray drying
like sedimentation, creaming and aggregation. Therefore, it is necessary conditions affects the characteristic of final product. Critical process
to preserve the original size of nanocrystals during their downstream parameters for this technique are inlet temperature, feed rate and as-
processing. Techniques such as freeze drying, spray drying, and spray- piration rate. Shegokar et al. studied the effect of different adsorbents
freeze drying are reported for the downstream processing of parenteral during spray drying. Sugars such as mannitol, lactose and maltodextrin
nanosuspension. were chosen as the adsorbents at 2–8% w/w concentration. Spray
drying with maltodextrin and mannitol yielded spherical particles while
4.1. Freeze drying lactose produced irregular shaped particles. The pH of the reconstituted
powder was around 7.01–7.11. Lower concentrations of 2% w/w of the
Lyophilization or freeze drying is commonly used technique for the adsorbents resulted in sticky powder. Powder with good flowability was
solidification of nanosuspension. It comprises of freezing, primary attained for lactose at higher concentrations of 4–8% during spray
drying and secondary drying steps which leads to the final moisture drying (Shegokar and Singh, 2012). In another study, researchers re-
content of the formulation below 1%. This dry powder can then be ported that sugars with lower glass transition (Tg) temperatures like
reconstituted immediately before use to generate nanosuspension and dextrose and sucrose resulted in sticky powders. On the other hand,
can be administered intravenously. Cryoprotectants are added into the lactose and mannitol (Tg > 80 °C) yielded flowable powder. Therefore,
formulations to protect the actives from freezing damage. Optimization it is suggested that Tg is a better predictor for the selection of ad-
of the cryoprotectant type and its concentration is important as it af- sorbents in comparison with melting point for spray drying perfor-
fects the final product quality. Shegokar et.al. investigated dextrose, mance (Chaubal and Popescu, 2008).
lactose, trehalose, mannitol, sorbitol, polyvinyl pyrrolidone, fructose, Aggregation of particles may be caused due to the excess stress
sucrose, glucose and reported that their protective effect is concentra- during the process of drying. Another reason causing aggregation of
tion dependant with lower concentrations yielding a sticky product. particles during spray drying include force of air, velocity, and contact
Hence it was suggested that the 6–8% concentration of the sugars time between particles (Gianfrancesco et al., 2008). Chaubal et al. in-
should be used to act as an effective protectant. The effect of ster- vestigated effect of spray drying on itraconazole nanosuspension. They
ilization process should also be assessed. During the process of reported that the mean particle size of spray dried particles was

5
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

10–20% higher than the mean of the original suspension. It was ob- and hence indicates short-term stability (Li et al., 2018). However, this
served that polymeric surfactants and sugars were insufficient to sta- may not be the essential requirement for stability. Nanosuspensions are
bilize the nanoparticles during the spray drying process and ad- reported to be stable without having zeta potential in the above men-
ditionally charged surfactant was needed to inhibit irreversible tioned range.(Verma et al., 2009b) This can be attributed to inherent
aggregation (Chaubal and Popescu, 2008). Hence process optimization differences in the density and effect of gravity between the nanocrys-
is necessary in order to generate redispersed nanosuspension for in- talline dispersions and other nanoparticulate systems.
travenous administration.

4.3. Spray freeze drying 5.2. Particle size and particle size distribution (PSD)

Spray freeze drying (SFD) technique is a combination of two pro- Mean particle size and PSD are important parameters as they control
cesses mentioned above. This technique involves three stages: droplet dissolution velocity, saturation solubility, physical stability and in-vivo
generation, freezing and sublimation drying. Droplets are generated by behaviour of nanosuspension. Photon correlation spectroscopy (PCS)
passing the nanosuspension through a nozzle and they are immediately can be used to determine the mean particle diameter and the range of
frozen in liquid nitrogen. These frozen droplets are now subjected to particle size distribution (polydispersity index, PI) of nanosuspension.
lyophilization and water is removed by sublimation drying (Niwa and The PI is significant as it dictates the physical stability of nano-for-
Danjo, 2013). SFD is particularly used for thermosensitive products like mulations and should be low enough for long-term stability of nano-
recombinant proteins and vaccines. This technique has been explored suspension. A PI value of < 0.3 indicates fairly narrow size distribution.
for injectables and inhalable products (Wanning et al., 2015). It is a Although PCS is a versatile and rapid technique, but it only covers the
promising approach for the downstream processing as it offers smaller range of 0.02–0.3 µm (Knapp et al., 1996). Hence, laser diffractrometry
particle size when compared to the spray dried product. However, there (LD) analysis should be performed additionally to PCS in order to detect
are no examples of drying IV nanosuspension using SFD. the drug microparticles that might be present in the formulation. LD
analysis becomes critical for IV nanosuspension as it determines a vo-
4.4. Electrospraying lume size distribution and can be used to measure particles in the range
of 0.05–80 µm. In some instruments particle sizes up to 2000 µm can be
Electrospraying, also called electrohydrodynamic atomization measured (Keck and Müller, 2008). This is important for IV nanosus-
(EHDA), allows generation of submicron droplets with a narrow size pension as size > 6 µm can lead to capillary blockage or emboli for-
distribution. It works on electromechanical and hydrodynamic forces mation. Another technique that can be employed for determination of
that function in combination with each other. Nanosuspension is passed particle size is the Coulter counter technique. The Coulter counter
through an emitter (glass or metal capillary) to which high voltage is provides the absolute number of particles per volume unit for the dif-
applied. Under the influence of electric field, the surface tension of li- ferent size classes in the range of 0.4 µm – 1600 µm (Müller and Peters,
quid causes the liquid jet to break into droplets. These droplets, when 1998). Hence, it is a more appropriate and efficient technique than LD
coupled with drying gas, will evaporate to form dry solid particles. analysis for measuring the microparticulate drug crystals present in
Voltage applied, flow rate and distance between tip to collector are nanosuspension. Both PCS and LD require proper dilution either with
critical parameters of this process (Chan and Kwok, 2011). Thakkar deionized water or stabilizer solution before the analysis to prevent
et al. made a comparative evaluation between electrospraying and multiple scattering, thus modifying the nature of nanosuspension to a
freeze drying as solidification techniques for erlotinib nanosuspension. certain degree (Li et al., 2015).
On the basis of various parameters like amount of drug, particle size, Visual characterization techniques like scanning electron micro-
morphology, solid state form of drug, surface area, pore volume, size scopy (SEM) and transmission electron microscopy (TEM) can be used
after redispersion and drug release, it was reported that lyophilized as supportive technique for the identification of primary drug nano-
powder was superior over electrospray dried powder. However, this crystals (Bilgili et al., 2016). With combined use of SEM/ TEM imaging
technique was investigated for preparing a nano spray dried powder for and particle sizing, the aggregation nature of nanosuspension can be
orodispersible tablets (Thakkar et al., 2018). It would be interesting to assessed qualitatively and quantitatively (Li et al., 2018). Due to the use
explore this technique further for injectables. of high vacuum in these techniques, water evaporation takes place
which obstructs the electron beam and interfere with the image re-
5. Characterization of nanosuspension solution.
Other techniques for particle size determination are wide angle X-
5.1. Zeta potential ray scattering (WAXS), small-angle X-ray scattering (SAXS) and X-ray
diffraction (XRD). These are indirect methods and need dry powder
Zeta potential (particle charge) provides an indication about the sample for analysis, but they offer more reliable information from the
physical stability of nanosuspension formulation. Zeta potential can be statistical viewpoint. XRD can be used for determining the particle size
determined by Zetasizer and a value of at least ± 30 mV is essential for from the widening of the XRD reflections by means of the Scherrer
an electrostatically stabilized nanosuspension (Müller and Hildebrand, equation, Williamson- Hall plot and Rietveld method. The Scherrer
1996). Indomethacine nanosuspension, stabilized with sodium lauryl equation is given by (Scherrer, 1918)
sulfate (electrostatic stabilizer), exhibited higher zeta potential values
ranging from −84 mV to −90 mV whereas lower zeta potential values K
D=
were found in the range of −17 mV to −20 mV when HPMC (steric cos
stabilizer) was used (Verma et al., 2011). In the case of collective sta-
bilization (by electrostatic and steric), a minimum zeta potential value where D is the particle size, K is a constant which depends on
of ± 20 mV is desirable. In one of the studies, authors investigated crystallite size (close to unity), λ is the wavelength of the radiation, β is
nanosuspensions that were stabilized through a combination of an the full width on a 2θ scale at half maximum intensity and θ is the
electrostatic stabilizer, soya lecithin and a steric stabilizer, tyloxapol. Bragg’s diffraction angle. In this method, average crystallite size has
The zeta potential of budesonide nanosuspension which combination of been calculated using Gaussian fit by fitting the XRD pattern. According
stabilizers was found to be −41.1 mV. (Jacobs and Müller, 2002). to Williamson Hall method, (Williamson and Hall, 1953), the individual
Values ranging from −5 to + 5 mV point towards faster aggregation contributions to the reflection broadening can be expressed as

6
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

2006).
cos = K + [4 sin ]
D
5.4. Solid state characterization
where 4ε sinθ is the effect of strain on the crystallites. This indirect
method of analysis is preferred when higher angle reflections are weak Process parameters used during generation of nanocrystals may
and difficult to analyze. The expression used in Rietveld method is cause changes in solid form of crystalline particles. Therefore, the final
expressed as (Young, 1993) solid form of drug in the formulation needs to be confirmed as it may
IG affect stability and its performance after IV administration. It was ob-
FWHM 2 = (U + DST 2)(tan2 ) + V (tan ) + W +
cos 2 served that during the production of nanosuspension, application of
high pressures led to amorphization of API, e.g. the drug RMKP22 was
where U, V and W are the usual peak shape parameters; IG is a found in amorphous state in the nanosuspension (Grau and Müller,
measure of the isotropic size effect and DST is the coefficient related to 1998). Differential scanning calorimetry (DSC) and XRD analysis are
strain. Particle size of nanoparticles can also be done using surface area most commonly used to identify the solid form of drug nanocrystals
analysis from BET which is a relatively simple technique to calculate (Liu et al., 2018). Percent crystallinity can be determined through XRD
the particle size. However, its limitation is that it is based on the hy- by plotting the calibration curve of different fractions of physical
pothesis of spherical shape of particles for calculation which is not mixtures containing crystalline and amorphous drug. It is imperative to
applicable to the various types of nanoparticles (Suri et al., 2016). More note that sample preparation is essential to determine the solid form of
recently, solid-state NMR spectroscopy (ssNMR) has been explored for API in the final formulation. Downstream processing of the nanosus-
the estimation of particle size in the nanoformulation. Munson et al. pension involves inclusion of additional excipients which may interfere
have investigated the correlation between particle size and ssNMR with the characterization process. Thermal events of DSC may overlap
proton spin–lattice relaxation (1H T1) times. They observed that smaller with that of the excipients used or excipient-drug interaction might lead
particles of dicumarol usually had shorter relaxation times than larger to polymorphic transition. Hence, it is advisable to carry out solid state
ones. This technique can be used to determine particle size of APIs even characterization of the API alone. This can be implemented by drying
in presence of excipients and such evaluation of particle size distribu- the nanosuspension formulation at ambient conditions. Critical para-
tion of the API could be helpful during the process of development. meters during sample preparation are drying temperature and drying
However, authors propose that there may be other factors that can be time. Care should be taken that drying temperature or the duration of
attributed to the change in relaxation time which needs to be explored the process do not affect the solid form generated during the manu-
to employ ssNMR as a tool for particle size evaluation (Dempah et al., facturing process. Additionally, electron microscopy can be used to
2017). Also on-line monitoring of size of particle is now possible with characterize the crystalline drug from the amorphous. Crystalline drug
the help of near-infrared (near-IR) spectroscopy. This technique offers appears bright against black polarized background in the birefringence
real-time data for process monitoring with an accuracy of 2.4 nm close mode in contrast to amorphous that appears dark. In conjugation with
to the endpoint of particle production (Higgins et al., 2003). In con- DSC, hot stage microscopy (HSM) can help in identifying events asso-
clusion, the particle size of nanoparticles needs to be confirmed using ciated with various thermal events aids differentiation of DSC en-
complementary tools to ensure the particle size in the given formula- dotherms for polymorphic transitions.
tion.
5.5. In vivo performance
5.3. Particle morphology
Determination of an in vivo performance of the formulation is an
Microscopy is the only technique where the distinct particles can be essential part of the study as it helps in the establishment of an in vitro\
visually detected, thus providing information about the morphology in vivo correlation (IVIVC). In vivo performance is currently evaluated in
besides particle size. Optical microscopy cannot be used in case of costlier, time-consuming rodent models. Utilization of the chick chor-
particles which are shorter than the wavelength of visible light. Hence, ioallantoic membrane (CAM) model is a rapid, accessible and low-cost
electron microscopy technique must be used for particle size in the alternative approach (Leong et al., 2010). CAM is highly vascularized,
nanometer range. Both SEM/ TEM techniques provide images with mimicking the diverging/converging vasculature of mammalian organs
highest resolution and can reveal the finest details of the structure such as the liver or spleen and is known to trap nanoparticles. In one
(Bilgili and Afolabi, 2012; Borchert et al., 2005). The sample must be study, CAM imaging revealed dramatically different circulation beha-
completely dry before SEM/ TEM analysis and so both SEM and TEM viours of colloidally stable cationic particles with identical size, shape
require proper sample preparation prior to analysis. The first step is and zeta potential, differing only in charge distribution (Townson et al.,
drying the sample which can be done by either freeze drying or freeze 2013). Nanoparticles with patchy charge were immediately seques-
fracture. In case of SEM analysis, the dry sample is made conductive by tered, whereas uniformly charged nanoparticles remained in circula-
coating the surface with gold, palladium, platinum or osmium. For TEM tion. This observation was later verified within a rodent model via
analysis, a coating of osmium tetraoxide is applied onto the dry sample SPECT imaging (Dogra et al., 2018). Importantly, the CAM model can
to provide sufficient contrast (Müller-Goymann, 2004). However, water also be utilized for nanoparticle–tumour interaction studies (Cho et al.,
loss may lead to changes in the microstructure of the sample and also 2011; Durfee et al., 2016). While rodent models remain indispensable
drying alters the existing aggregation state of drug nanosuspension (Li for new investigational drugs, the CAM model assertively assists as an
et al., 2018). Apart from this, rheological characterization of nanosus- inexpensive, efficient intermediary system which can help qualify na-
pension could support the assessment of the aggregation state (Azad nosystems for subsequent mammalian testing. This will also reduce the
et al., 2015). Atomic force microscopy works on the principle of scan- number of mammalian animals utilized and help bridge the in vitro to in
ning probe microscopy and also reflects the surface topography of vivo gap (Leong et al., 2019). The in vivo behaviour of the drug is in-
sample (Verma et al., 2009a). Sample preparation for AFM is simple fluenced by its surface hydrophobicity and interactions with plasma
than SEM/ TEM and the samples do not have to be conductive. The proteins. The composition of protein adsorption pattern obtained after
nanocrystalline particles are to be dispersed or adhered on a substrate the IV injection of nanocrystals is considered as a crucial factor for
with the help of an adhesive in order to characterize them. Most organ distribution (Blunk et al., 1993; Blunk et al., 1996; Lück et al.,
commonly used substrates are mica, glass cover slips, highly oriented 1998). Therefore, appropriate techniques must be employed so that the
pyrolytic graphite (HOPG), atomically flat gold and silicon oxide wafers protein interactions and surface properties can be evaluated. Surface
and poly-L-Lysine is the commonly used adhesive (Starostina and West, hydrophobicity can be determined by hydrophobic interaction

7
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

chromatography (Wallis and Müller, 1993) whereas 2-D PAGE (Blunk and number of homogenization cycles (Al Shaal et al., 2010). It is
et al., 1993) can be utilized for the measurement of protein adsorption usually recommended to start with micronized suspension for both HPH
both qualitatively and quantitatively after IV injection of drug nano- and WMM processes since clogging of the equipment can occur when
suspensions. the process is steered with coarse suspension. All combination tech-
nologies mentioned earlier can perform better than standard WMM and
6. Scale-up HPH, however, any pre-treatment step would increase the complexity
of the entire process and can add to the overall cost (Möschwitzer,
Drug nanocrystals are incorporated in the formulation development 2013).
decision trees of several pharmaceutical companies as they are amen-
able to scalability. Scalability is achieved by the sameness condition,
the similarity among laboratory, pilot and the production. Majority of 7. Regulatory aspects
the drug products available in the market have been prepared using
WMM and HPH due to the ease of scalability. In case of WMM, process The US Food and Drug Administration (FDA) regulates nano-
parameters and material attributes like the amount of the drug, amount technology products within the existing statutory legal standards ap-
of milling pearls used, milling speed, milling time and temperature plicable to each type of product. Technical assessments are made by the
must be controlled to obtain particles in the desired size range. WMM is agency on the basis of the product and science-based policy, thus al-
preferred due to the availability of suitable equipment like agitated ball lowing tailored-made approaches that reveal the characteristics of
mills where the drug is milled for about 30–120 min to obtain nano- particular products (e.g., nanocrystals), besides incorporating evolving
crystals of desired size. They can operate in batch mode or continuous technology and scientific understanding within the field. In 2014, FDA
mode (recirculation mode). For large scale operation it is often used in released a guidance document for industry that describes the agency’s
recirculation mode to obtain the desired particle size. These agitated thinking on considering whether FDA- regulated products involve the
mills have media separators like filter cartridge or separating gap application of nanotechnology. This guidance document considers two
system that allows it to hold the milling media back in the milling points in evaluating if the product involves application of nano-
chamber while the nanosuspension is circulating. The suspension is technology: (1) a material or end product is generated to have at least
pumped back into the milling chamber with a certain velocity and one external or an internal dimension or structure in the nanometer
hence within a short residence time in the chamber, the particles are range (approximately 1 to 100 nm), (2) an end product is engineered to
exposed to high energy input thereby reducing its particle size (Kwade, unveil physical or chemical properties or biological effects that are
1999). Planetary ball mills with modified sample holders are available attributable to its dimensions up to 1000 nm.
for small-scale production of nanosuspension. Alternatively, agitated Generic products containing nanocrystal APIs can be submitted to
ball mills are used for drug quantities starting from 10 mg (Merisko- the FDA for approval through 505 (b) (2) route that allows the appli-
Liversidge and Liversidge, 2011). Using these mills, it is now possible to cant to rely on the studies or data previously available in the original
produce nanosuspensions during the early discovery phase of the for- application or published in the literature. For example, Ryanodex®
mulation development or to perform stabilizer screening studies with a (dantrolene sodium) is the nanocrystalline product which is a re-
minimal API consumption. WMM can be viewed as a scalable approach formulated product from Dantrium Intravenous, a lyophilized for-
with the availability of appropriate equipment for small scale to com- mulation (micronized) for injection. One of the reports revealed that
mercial production batch. submissions for nanocrystals comprises of approximately 30% of all the
HPH is another technique that is commonly used to produce na- applications for drug products containing nanomaterials (Chen et al.,
nocrystals. High pressure homogenizers are commonly available in the 2017). Nanocrystalline suspension for oral or intravenous administra-
pharmaceutical industry and hence can be utilized in the commercial tion are generated through the top-down and bottom-up techniques
production of nanosuspension. HPH is unlikely to generate impurities mentioned earlier. Hence, their critical quality attributes (CQAs) gov-
compared to WMM as a consequence of abrasion and wearing of the erning the quality remain the same.
equipment. A comparative study revealed that a typical nanosuspension A nanocrystalline drug product specification includes the CQAs
after 20 cycles at 1500 bar contained < 1 ppm iron (Krause et al., encompassing PSD, zeta potential, particle shape, polymorphism, drug
2000). However, in piston-gap homogenizers, wearing and abrasion can content and drug release.(Chen et al., 2017; Peltonen, 2018) Several
arise when very hard material is processed. This happens because the process variables like critical material attributes (CMAs) and critical
tip of the homogenization valve consists of a comparatively small sur- process parameters (CPPs) must be controlled with the aim of achieving
face in comparison to the volume of suspension moving across it. They the desired CQAs. Examples of CQAs for drug nanocrystals and how
become worn out when stainless steel parts are used thereby affecting they are affected by various process variables like CPPs and CMAs are
the process efficiency. Therefore, modern homogenizers have valves captured in Table 4. Various quality considerations from chemistry,
fitted with ceramic tips, providing additional protection in harsh con- manufacturing and controls (CMC) perspective also includes control of
ditions making it more suitable for parenteral use (Innings et al., 2011). drug substance and drug product and their stability. It is important to
In HPH, the coarser suspension is passed several times (i.e. homo- include crystallinity as well as PSD in the drug substance specifications
genization cycles) to achieve the desired particle size. In order to avoid for nanocrystals. The agency proposes to subject the products (invol-
clogging of the narrow homogenization gap, the applied pressure is ving application of nanotechnology) to premarket review. In case the
increased from 10% to 100%. Production pressure usually spans be- products are not subjected to premarket review process, the FDA urges
tween 1000 and 2000 bar wherein the opening gap is only of a few the industry to consult the agency in the initial stage of product de-
micrometer (Möschwitzer, 2013). There are reports that describe de- velopment.
creased microorganism load and enzyme inactivation as a result of HPH The European Nanomedicine Characterisation Laboratory (EUNCL)
process (Dumay et al., 2013). Today, homogenizers are available from and the REFINE consortium effort, funded by EC-H2020, are aimed at
mL-scale to commercial production scale, making HPH a scalable pro- developing a regulatory science framework for nanomedicine. EUNCL/
cess (Keck and Müller, 2006). In one of the studies, a comparative REFINE, jointly with the National Cancer Institute’s Nanotechnology
evaluation was made between product produced at lab scale and pilot Characterization Lab (NCI-NCL), would bridge the gap between pub-
scale using WMM and HPH. The authors concluded that the particle size lication and translation by identifying common pitfalls in nanomedicine
of the nanosuspension was dependent on the velocity of pumping, development. It would also define critical quality attributes for pre-
agitator rotation speed and the number of milling cycles. Particle size clinical assessment thus enabling product development (Caputo et al.,
reduction by HPH was observed to be affected by the applied pressure 2019; Crist et al., 2013; Gioria et al., 2018; Swierczewska et al., 2018).

8
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

Table 4
Example for risk assessment matrix for drug nanocrystals analyzing the impact of CMAs and CPPs on CQAs (Peltonen, 2018).
Risk assessment matrix
CQAs CMAs CPPs
Stabilizer type Stabilizer concentration Drug amount Milling time Milling speed Bead size

Particle size Medium Medium Medium High High High


PDI Medium High Medium High High High
Zeta potential High High Medium Medium Medium Medium
Drug content Low Low Low Low Low Low
Drug release Medium Medium Medium High High High

8. Applications the mixture, there is reduced contamination and interruption amongst


the runs. This process has been developed by ResodynTM corporation
8.1. Role in preclinical formulations with their ResonantAcoustic® Mixing method and various commercial
acoustic mixers are available.
Preclinical formulations are an integral part of drug discovery and
development. Their development follows both the stages of drug de-
8.2. Targeting of nanosuspension
velopment i.e. at discovery stage (lead discovery) and at preclinical
stages (preclinical lead) (Li and Zhao, 2007). These early formulations
Nanocrystals have recently emerged as a formulation strategy to
help to optimize in vivo exposure, predict initial dosing for human
target drugs at specific site of action for a better therapeutic effect.
clinical trials and reduce toxicological side effects. General tox-
Nanoparticulate systems have revealed great potential in delivery of
icological (tox) studies are carried out to assess the adverse effects of
drugs to the targeted organs of the body (Chen et al., 2018). Similarly,
investigational compounds in vivo. Tox formulations present significant
nanocrystalline suspension can be used for targeting of drugs due to
challenges to a formulation scientist as these formulations demand high
their surface potential and their in vivo behaviour.
concentrations and they are used for a long time period (Li and Zhao,
Following IV injection, drug nanocrystals can extravasate to the
2007). For example, in dose escalation studies, it is necessary that the
tumors from the leaky endothelium via the enhanced permeation and
formulation delivers numerous folds greater in vivo exposure than the
retention effect (EPR). This phenomenon is known as passive targeting
effective dose and also the preparation is dosed incessantly for definite
which is mainly a result the submicron size of nanocrystalline particles
period of time (for example, 2 weeks). It is important to note that the
(Maeda, 2015). Nanocrystals may behave analogous to the drug solu-
vehicle is anticipated not to provoke any adverse effect. Toxicological
tion after IV administration due to rapid dissolution profile achieved in
studies are critical for a lead compound on its way to progress. Dis-
in vivo sink conditions (Mouton et al., 2006). If their dissolution is not
covery leads usually are in limited quantity (for example, < 20–30 mg)
immediate then, they are taken up by macrophages predominantly in
and hence the early formulation development is a challenge if the drug
the liver (Kupffer cells) and spleen, later diffusing slowing across the
compound has poor solubility.
concentration gradient (Rabinow, 2004). This IV depot action of the
Preclinical leads require detailed efficacy/activity, pharmacokinetic
formulation offers a pharmacokinetic profile with prolonged t1/2 and
and toxicological profile. Requirement of a formulation selection is
significantly reduced Cmax. This behaviour of nanocrystals is beneficial
more specific here. Nanosuspensions can serve as an immediate solu-
for some classes of drug, for whom toxicity is intervened by peak
tion for the preclinical formulations as they provide high drug con-
plasma values, nonetheless the efficacy is determined by AUC as ob-
centrations and are devoid of a large number of excipients (Kesisoglou
served in the event of antifungals (Andes, 2003). In a clinical trial
and Mitra, 2012). Nanosuspension for early formulations can be de-
(Donnelly et al., 2001), the pharmacokinetic parameters of nanocrys-
veloped using the NanoMill® (< 50 mL) or high pressure homogenizers
talline suspension of itraconazole were compared to the commercial
(provided they handle small volumes efficiently). In one of the case
Sporanox solution. The nanosuspension with particle size of about
studies, it was reported that a neutral compound having 2 µg/mL as its
200–300 nm employed a larger volume of distribution (1,677 ± 827 l
solubility and a log P of 2.5 was to be tested in monkeys. The drug
versus 796 ± 185 l) and a slow clearance (3.35 ± 1.8 l/h versus
compound was not adequately lipidic to be incorporated in a lipidic
22.9 ± 5.7 l/h) to provide a longer t1/2 (346 ± 225 h terminal versus
system and solubilizing agents like Captisol®, vitamin E TPGS, HPβCD
35.4 ± 29.4 h mean and 30 h terminal) and larger area under the
proved to be futile to solubilize the drug. Hence, a nanosuspension was
plasma concentration curve for the initial 24 h, AUC24,
developed using the microfluidization technique which generated a
(51,558 ± 10,635 (g•h/l versus 30,605 ± 8,961 (g•h/l) (Willems
particle size (D50) of 40 nm. When such nanosuspension formulation
et al., 2001). The study presented above confirms the mononuclear
was administered to monkeys, it attained 130% of the exposure of the
phagocyte system (MPS) depot behaviour, resulting in delivery for
standard suspension (Boersen et al., 2013). There are reports available
itraconazole nanosuspension for a prolonged period of time. In another
in the literature where nanosuspension have been successfully devel-
finding, it was reported that itraconazole nanosuspension having an
oped for preclinical studies. For example, a poorly water soluble com-
average particle size of 268.1 ± 6.5 nm showed similar dissolution
pound, 1,3- Diclyclohexylurea (DCU), which lowers blood pressure was
profile to that of marketed itraconazole injections. However, it dis-
developed as nanosuspension for infusion and IV bolus dosing
played different pharmacokinectic properties such as decreased initial
(Wahlstrom et al., 2007). In another study, nanosuspension for in-
drug concentration, increased mean residence time (MRT) and plasma
travenous administration in preclinical studies of a poorly aqueous
half-life, and increased concentration in the liver, spleen, and lung in
soluble compound BI XX was developed using ball mill (Frank and
comparison to commercial itraconazole injections. Therefore, nano-
Boeck, 2016).
suspension could change the in vivo distribution of itraconazole also
A novel drug sparing technology employing acoustic mixing is re-
helping in passive targeting to MPS (Yuan et al., 2019)
ported to identify nanosuspension formulations for widespread com-
In one of the studies, it was reported that clofazimine nanosuspen-
pounds with enhanced efficiencies (Leung et al., 2014). This technique
sion (particle size 385 nm) administered intravenously demonstrated
applies high intensity acoustic energy coupled with low frequency to
targeting to the reticuloendothelial system (RES), successfully targeting
the mixer with low shear to generate nanosuspension. Since the mixer
the drug to major host cells in mycobacterial infections i.e. tissue
agitates the whole container and has no contact with the components of
macrophages. Since clofazimine has low solubility in water (0.3 g/l at

9
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

pH 7.8), these nanocrystals did not dissolve immediately and resulted in of pegylation, stabilization and CD44 receptor targeting. It was ob-
the sequestration of nanocrystals in the RES (Peters et al., 2000). Ganta served that in MDA-MB-231 cells, DTX-CSA NCs exhibited enhanced
et.al. reported that asulacrine (ASL) nanosuspension with average size, cellular uptake, deeper penetration and exhibited higher degree of cy-
133 ± 20 nm, was rapidly cleared from the plasma and showed re- totoxicity. The authors concluded that this was due to the EPR effect
markably increased concentrations in tissues like the liver, lungs and combined with receptor mediated endocytosis. IV administration of
kidney. The authors have rationalized that this was due to the uptake of DTX-CSA formulation demonstrated improved pharmacokinetic profile
the nanocrystals by the MPS which cleared them from systemic circu- and significant inhibition in 4T1 induced tumor with reduced toxicity
lation. Therefore, ASL nanosuspension had a distinctly larger AUC0-∞ in (Pandey et al., 2018).
kidney and liver but not in heart (Ganta et al., 2009). Gao et al. have
studied particle size effect on the tissue distribution and pharmacoki- 8.2.1. Challenges due to shedding of ligands
netics of two oridonin NCs with distinctly different sizes Stabilizers and targeting ligands are non-covalently linked onto the
(103.3 ± 1.5 nm and 897.2 ± 14.2 nm) following IV administration surface of nanocrystals and nonspecific interactions like adsorption are
in rabbits. Complete dissolution was observed in vitro for the smaller predominant during surface modification. A weak interaction is pro-
and larger NCs in 10 min and 2 h, respectively. In vivo, the smaller NCs vided by physical adsorption and leads to desorption of stabilizer
performed in the same way as drug solution, while the larger NCs ac- during the dissolution of nanocrystals. Excessive dilutions faced during
cumulated to a greater degree in the liver, lungs and spleen. The au- in vitro or in vivo experiments may result in shedding of the stabilizer or
thors proposed that the small crystals dissolved completely in short targeting ligands. For example, Deng et al. have demonstrated with the
span of time in vivo thus behaving like solution whereas the larger NCs help of increased crystal size that poloxamer 407 (Pluronic® F127)
remained insoluble and were subjected to MPS uptake (Gao et al., detached from paclitaxel nanocrystals on dilution or mild heating
2008b). This is in line with the studies discussed earlier. In another (Deng et al., 2010). Therefore, shedding of ligands/ stabilizers is a
study, Zhang et al. demonstrated a superior antitumor efficacy for un- critical obstacle when the aim is to offer targeting to a particular site.
coated camptothecin NCs having a particle size of 240 nm in a MCF- 7 Chemically altering the stabilizer to provide more sites for interac-
tumor xenograft mouse model in comparison to the camptothecin salt tion with nanocrystals can be one of the approaches to reduce shedding.
solution made of propylene glycol and saline. This was ascribed to the Another approach includes crosslinking the stabilizer around drug na-
EPR effect and higher hydrolysis resistance of NCs. They additionally nocrystals thereby reducing shedding via physical entrapment and
supported their observations with biodistribution data presenting physisorption. For instance, Fuhrmann et al. crosslinked block copo-
greater deposition of camptothecin in the tumor and reduced drug lymer directly on the exterior of PTX NCs to form polymeric nanocages.
hydrolysis (Zhang et al., 2011). It was reported that these nanocages-NCs provided steric barrier to
It is evident from the aforementioned studies that altering the par- particle–particle interaction and prevented aggregation. Size-stability
ticle size plays an important role in targeting of nanocrystals. Smaller analysis revealed that the nanocages-NCs had better size-stability when
crystals target tumour cells by virtue of their EPR effect and larger compared with non-cross-linked coating. It was demonstrated that the
crystals are up taken by MPS predominantly in the spleen and liver. nanocages showed 3 to 4 times less shedding from the NCs surface
Apart from this, targeting ligands can be used for surface mod- which was achieved by tailoring the extent of polymer release from the
ification of drug nanocrystals. Surface modification will also help to nanocage-NCs. Transmission electron microscopic images obtained
prolong the in vivo dissolution of nanocrystals. Sharma et al. in- after dissolution of NCs were evident that the nanocages remained in-
vestigated hyaluronic acid (HA) anchored paclitaxel (PTX) nanocrystals tact (Fuhrmann et al., 2012). Kim and Lee have reported crosslinking of
(HA-PTX/NC) for its chemotherapeutic efficacy. It was observed that chitosan by tripolyphospate on the surface of PTX and naproxen. The
HA-PTX/NC markedly prolonged the systemic circulation of PTX and crosslinking significantly modified the release profiles of the NCs.
increased the AUC0-∞ by 8.4 times in comparison with marketed for- However, they did not evaluate the size stability and shedding phe-
mulation of PTX i.e. TaxolTM. The study also revealed that HA-PTX/NC nomenon after crosslinking (Kim and Lee, 2011).
exhibited reduced lung metastatis, greater antitumor efficacy, and less More recently, the Layer-by-Layer (LbL) assembly of polyelec-
toxicity in LA-7 tumor bearing rat model in comparison to TaxolTM trolytes has been explored as an approach to obtain stabilized coatings
(Sharma et al., 2016). In another study, PTX-NC, surface modification for the encapsulation of drug NCs. In this approach, first step is to an-
with HA and apo-transferrin (Tf), was assessed for their inhibition of chor a layer of small amphiphile molecule and a polymer to the hy-
cell growth. In vitro results presented higher cell permeability in con- drophobic drug NC tailed by deposition of many layers of charged
trast to the unchanged PTX-NC. Also in MCF-7 cells the surface mod- polyelectrolytes sequentially. The drug release is prolonged due to the
ification of PTX-NCs led to 60% cell growth inhibition, but the effect thickness of the polymer layers which controls the diffusion of drug out
was inferior than unmodified PTX-NC and pure PTX in normal cell line of the encapsulation. Polomska et al. adopted the LbL approach to coat
(Sohn et al., 2017). Noh et al. prepared herceptin (HCT) functionalised PTX NCs with alternating layers of oppositely charged polyelectrolytes
PTX-NCs and studied their effect on HER-2 positive breast cancer cells. and the top coat with a PEGylated copolymer. It was observed that
The cell uptake studies revealed that surface modified nanocrystals dissolution of coated particles was slower than the noncoated NCs and
exhibited higher binding affinity and greater cell-specific uptake to Abraxane (marketed formulation of PTX). However, the pharmacoki-
HER-2 positive breast cancer cell lines than PTX-NCs (Noh et al., 2016). netic and biodistribution profiles indicated that the coated NCs were
Shubar et al. investigated the role of apolipoprotein E (apo E) coat on rapidly removed from the blood stream. The authors hypothesized that
the surface of atovaquone nanocrystals (ANCs). It was observed that this was due to the shedding of PEGylated coating from the surface of
incubation of apo E coated ANCs with brain endothelial cells resulted in NCs (Polomska et al., 2017).
the accumulation of nanoparticles on the surface but not in their uptake In some cases, this shedding of stabilizers may be beneficial. For
into the brain cells (Shubar et al., 2009). Shegokar et al. developed instance, Liu et al. studied the shedding phenomenon by preparing PTX
nevirapine nanosuspension with surface modification by dextran, NCs stabilized with D-α-tocopheryl polyethylene gycol 1000 succinate
serum albumin, and polyethylene glycol to improve its targeting action. (TPGS). The rationale of this study was that TPGS acts as a P-gp in-
In vivo tissue distribution studies showed an increased NCs accumula- hibitor which on shedding improved the drug uptake in resistant cells.
tion in various organs like brain, liver and spleen thereby prolonging This was indeed demonstrated that in NCI/ADR-RES cells, over-
their residence at target site as compared to pure drug (Shegokar and expressing P-gp and PTX resistant, TPGS stabilized formulation pre-
Singh, 2011). Pandey et al. prepared docetaxel (DTX) nanocrystals and sented a distinctly improved anti-proliferative effect in comparison to
stabilized them with a novel chondroitin sulphate A (CSA) and poly- free PTX or poloxamer 407 stabilized PTX formulation (Liu et al.,
ethylene glycol congujate. This stabilizer provided multiple advantages 2010).

10
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

9. Marketed and pipeline nanosuspension products for IV

Marketed

Phase 3
Phase 2
administration

Status
In case of IV delivery, after the approval of a nanoparticulate IV

Eagle Pharmaceuticals
suspension- Abraxane® in 2005, there has been a steady increase in the
drug particulate formulations moving into human clinical trials.

Pharma company
Abraxane® consists of paclitaxel protein-bound particles wherein pa-

Recro Pharma
clitaxel is entrapped in an albumin matrix. However, paclitaxel exists in

Berg, LLC
a non-crystalline i.e. amorphous state in Abraxane® which is different
than the nanocrystal-based nanosuspensions discussed here. Drug na-
nocrystals are one of the most successful formulations that have wit-

technology
nessed short time frame between inventions to market stage. However,
this is true for oral and other routes of administration. Few products

Applied Technology
have now reached clinical phases and only one product is available in

TM
the market for IV administration. This may be due to challenges with

NanoCrystals
the translation development, sterility and long-term stability of these
formulations. Ryanodex® is the only commercial product for IV route. It
is a lyophilized formulation of dantrolene sodium which is recon-


stituted with sterile water for injection to form nanosuspension. Apart

Mannitol, polysorbate 80, povidone K12, sodium hydroxide or hydrochloric acid


from this, meloxicam nanocrystals for IV route have reached in the late
phase of clinical trials (phase 3) and ubidecaronone has reached phase
2 clinical trials. Table 5 summarizes the marketed and pipeline nano-
suspension products for IV administration and Table 6 consists of re-
ported nanosuspension for IV administration in the literature.

10. Future perspective

Advancements in formulation approaches and commercially fea-


sible manufacturing processes have aided in bringing nanosuspension
from researcher's lab to patient's bed at an augmented rate. There are a
few nanosuspension based intramuscular (IM) injections already in the
market like Invega Sustenna (Janssen Pharmaceuticals) and
Aripiprazole Lauroxil (Alkermes, Inc.) and Aripiprazole long acting
injection (once a month) by Otsuka Pharmaceuticals used to treat
Schizophrenia. Long acting GSK1265744 (744) and rilpivirine
for pH adjustment.

(TMC278) nanosuspension achieved steady state plasma concentrations


Excipients used

within 3 days which proved its potential use in HIV-1 treatment (Spreen
et al., 2014). This trend is likely to increase in the future as pharma-
ceutical companies are looking at creating nanosuspension based IM
formulations because of their unique advantages of ease of adminis-


Marketed and pipeline nanocrystalline suspension products for IV administration.

tration and reduction in irritation or pain at the injection site. They are
Acute post-operative pain
Malignant hyperthermia

either depending upon the use of an oily medium or solvent or lipo-


philic prodrugs to obtain extended action. These formulations are
Pancreatic cancer
Therapeutic uses

especially useful in population that are susceptible to noncompliance of


dose e.g. patients with C.N.S. disorders like Schizophrenia. Similarly, in
other immunocompromised patients who have to take large number of
pills daily, it is going to offer a very convenient way of administration
of drugs. IV nanosuspension are pretty common formulation strategy in
the preclinical studies as they allow determination of IV pharmacoki-
netics. However, there are complication associated with the accumu-
lation of drug nanocrystals in the different organs like the RES systems
100 mg/kg (IV
1–2.5 mg/ kg

leading to organ toxicity. Unless these challenges are overcome, com-


mercial products seem to have a difficulty from a regulatory perspec-
infusion
30 mg

tive. At the same time, drugs that are sparingly soluble could be good
Dose

candidates for IV administration because they behave like IV solutions


after administration by virtue of their relative better pharmacokinetics.
Dantrolene sodium

Recent advances in aseptic technologies such as barrier isolation tech-


Ubidecaronone

nology aids in dealing with the technical hurdles of the aseptic pro-
Meloxicam

cessing requirements. Further the development of nanocrystals deco-


Drug

rated with functionalized surface coatings capable of targeting various


organs with high affinity can be viewed as the upcoming stage in the
nanosuspension research.
Trade Name

BPM31510
Ryanodex®
Table 5

N1539

11
D. Patel, et al.

Table 6
Reported nanosuspension for IV administration.
API Excipients Solvents Manufacturing technique Sterilization Critical parameters Particle size

Itraconazole Poloxamer 188 N-methyl- 2- Microcrystallization followed by Sterilized isolator was used for – 581 ± 18 nm
pyrrolidone HPH microcrytsallization
Omeprazole 1% Poloxamer 188 in 8.4% sodium bicarbonate – DissoCubes – Temperature, Number of 598–603 nm
homogenization cycles, Drug
content
Asulacrine Poloxamer 188 at 1% w/v – Pre homogenization using Ultra- – Temperature 133 ± 20 nm
turrax followed by HPH
Azoxystrobin 0.9% w/w of 1-Dodecanesulfonic acid sodium salt, – Wet media milling – Milling speed and time 238.1 ± 1.5 nm
polyvinylpyrrolidone K30
Camptothecin No additional stabilizers were added DMSO, Sonication with Antisolvent – 200–700 nm
Acetonitrile precipitation method
Curcumin 0.2% w/v Combination of soya lecithin and sodium – HPH – Number of homogenization cycles 132.6 to 360.8

12
deoxycholate and pressure
Clofazimine 0.5% Pluronic F68, 0.6% Phospholipon 90, 0.25% – HPH Aseptic conditions Number of homogenization cycles −385 nm LD (99) value-
Sodium cholic acid, 5.6% mannitol and pressure 2.28 μm
Tarazepide 1% w/w Poloxamer 188, 0.5% w/w Tween 80, Glycerol – HPH Production under laminar air flow Number of homogenization cycles 347–517 nm
as osmotic agent cabinet and pressure
Melarsoprol 1% Poloxamer 188 or 1% Poloxamer 407, 0.5% – HPH – Homogenization pressure, number 324 ± 88 to 663 ± 129 nm
mannitol of cycles
Docetaxel Chondroitin sulphate A-polyethylene glycol conjugate – HPH – Homogenization pressure, number 195.6 ± 12.5 nm
of cycles
Nevirapine 2.8% (w/w) of surfactant solution, Tween 80 (1%), – HPH – Homogenization pressure, number 468.9–520.3 nm
VolpoL4 (0.9%), Plasdone (0.1%), Poloxamer 188 of cycles
(0.5%), PVP (0.3%)
Oridonin 0.5% w/v Poloxamer and Lecithin (molar ratio 3:1), 1% – HPH – Homogenization pressure, number 103.3 ± 1.5 nm and
mannitol as cryoprotectant of cycles 897.2 ± 14.2 nm
Atovaquone 0.3% Tween 80, 0.3% Poloxamer 188 and 0.05% – High pressure homogenization Aseptic processing Homogenization pressure, number 279–286 nm
sodium cholate, glycerol as osmotic agent of cycles
Nimodipine 0.6% (w/v) poloxamer 188, 0.4% (w/v) sodium cholic – High pressure homogenization γ-ray radiation Homogenization pressure, number 300–700 nm
acid and 4.0% (w/v) mannitol of cycles
International Journal of Pharmaceutics 586 (2020) 119555
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

Declaration of Competing Interest Deng, J., Huang, L., Liu, F., 2010. Understanding the structure and stability of paclitaxel
nanocrystals. Int J Pharm. 390, 242–249.
Dogra, P., Adolphi, N.L., Wang, Z., Lin, Y.-S., Butler, K.S., Durfee, P.N., Croissant, J.G.,
The authors declare that they have no known competing financial Noureddine, A., Coker, E.N., Bearer, E.L., 2018. Establishing the effects of meso-
interests or personal relationships that could have appeared to influ- porous silica nanoparticle properties on in vivo disposition using imaging-based
pharmacokinetics. Nat. Commun. 9, 1–14.
ence the work reported in this paper. Dong, Y., Ng, W.K., Shen, S., Kim, S., Tan, R.B., 2009. Preparation and characterization of
spironolactone nanoparticles by antisolvent precipitation. Int. J. Pharm. 375, 84–88.
References Donnelly, J., Mouton, J., Rhijlevens, N., 2001. Pharmacokinetics of a 14 day course of
itraconazole nanocrystals given intravenously to allogeneic haematopoietic stem cell
transplant (HCST) recipients, 41st Intersci. Antimicrob. Agents Chemother. A, Conf.
Ahire, E., Thakkar, S., Darshanwad, M., Misra, M., 2018. Parenteral nanosuspensions: a Dumay, E., Chevalier-Lucia, D., Picart-Palmade, L., Benzaria, A., Gràcia-Julià, A., Blayo,
brief review from solubility enhancement to more novel and specific applications. C., 2013. Technological aspects and potential applications of (ultra) high-pressure
Acta Pharmaceutica Sinica B. homogenisation. Trends Food Sci. Technol. 31, 13–26.
Al Shaal, L., Müller, R., Shegokar, R., 2010. smartCrystal combination technology–scale Durfee, P.N., Lin, Y.-S., Dunphy, D.R., Muñiz, A.e.J., Butler, K.S., Humphrey, K.R., Lokke,
up from lab to pilot scale and long term stability. Pharmazie 65, 877–884. A.J., Agola, J.O., Chou, S.S., Chen, I.-M., 2016. Mesoporous silica nanoparticle-sup-
Andes, D., 2003. In vivo pharmacodynamics of antifungal drugs in treatment of candi- ported lipid bilayers (protocells) for active targeting and delivery to individual leu-
diasis. Antimicrob. Agents Chemother 47, 1179–1186. kemia cells. ACS nano 10, 8325-8345.
Azad, M., Afolabi, A., Bhakay, A., Leonardi, J., Davé, R., Bilgili, E., 2015. Enhanced Fagerberg, J.H., Tsinman, O., Sun, N., Tsinman, K., Avdeef, A., Bergström, C.A., 2010.
physical stabilization of fenofibrate nanosuspensions via wet co-milling with a su- Dissolution rate and apparent solubility of poorly soluble drugs in biorelevant dis-
perdisintegrant and an adsorbing polymer. Eur J Pharm Biopharm. 94, 372–385. solution media. Mol. Pharm. 7, 1419–1430.
Bergström, C.A., Charman, W.N., Porter, C.J., 2016. Computational prediction of for- Frank, K.J., Boeck, G., 2016. Development of a nanosuspension for iv administration:
mulation strategies for beyond-rule-of-5 compounds. Adv. Drug Deliv. Rev. 101, From miniscale screening to a freeze dried formulation. Eur. J. Pharm. Sci. 87,
6–21. 112–117.
Bilgili, E., Afolabi, A., 2012. A combined microhydrodynamics–polymer adsorption Fuhrmann, K., Schulz, J.D., Gauthier, M.A., Leroux, J.-C., 2012. PEG nanocages as non-
analysis for elucidation of the roles of stabilizers in wet stirred media milling. Int J sheddable stabilizers for drug nanocrystals. ACS nano 6, 1667–1676.
Pharm. 439, 193–206. Ganta, S., Paxton, J.W., Baguley, B.C., Garg, S., 2009. Formulation and pharmacokinetic
Bilgili, E., Li, M., Afolabi, A., 2016. Is the combination of cellulosic polymers and anionic evaluation of an asulacrine nanocrystalline suspension for intravenous delivery. Int.
surfactants a good strategy for ensuring physical stability of BCS Class II drug na- J. Pharm. 367, 179–186.
nosuspensions? Pharm Dev Technol. 21, 499–510. Gao, L., Zhang, D., Chen, M., 2008a. Drug nanocrystals for the formulation of poorly
Blunk, T., Hochstrasser, D.F., Sanchez, J.C., Müller, B.W., Müller, R.H., 1993. Colloidal soluble drugs and its application as a potential drug delivery system. J Nanopart Res
carriers for intravenous drug targeting: plasma protein adsorption patterns on sur- 10, 845–862.
face-modified latex particles evaluated by two-dimensional polyacrylamide gel Gao, L., Zhang, D., Chen, M., Duan, C., Dai, W., Jia, L., Zhao, W., 2008b. Studies on
electrophoresis. Electrophoresis 14, 1382–1387. pharmacokinetics and tissue distribution of oridonin nanosuspensions. Int. J. Pharm.
Blunk, T., Lück, M., Calvör, A., Hochstrasser, D., Sanchez, J.-C., Müller, B., Müller, R., 355, 321–327.
1996. Kinetics of plasma protein adsorption on model particles for controlled drug George, M., Ghosh, I., 2013. Identifying the correlation between drug/stabilizer proper-
delivery and drug targeting. Eur. J. Pharm. Biopharm. 42, 262–268. ties and critical quality attributes (CQAs) of nanosuspension formulation prepared by
Boedeker, B.H., Lojeski, E.W., Kline, M.D., Haynes, D.H., 1994. Ultra-Long-Duration Local wet media milling technology. Eur. J. Pharm. Sci. 48, 142–152.
Anesthesia Produced by Injection of Lecithin-Coated Tetracaine Microcrystals. J. Clin Gianfrancesco, A., Turchiuli, C., Dumoulin, E., 2008. Powder agglomeration during the
Pharmacol. 34, 699–702. spray-drying process: measurements of air properties. Dairy Sci Technol 88, 53–64.
Boersen, N., Lee, T., Hui, H.-W., 2013. Development of preclinical formulations for tox- Gioria, S., Caputo, F., Urbán, P., Maguire, C.M., Bremer-Hoffmann, S., Prina-Mello, A.,
icology studies, A Comprehensive Guide to Toxicology in Preclinical Drug Calzolai, L., Mehn, D., 2018. Are existing standard methods suitable for the evalua-
Development. Elsevier 69–86. tion of nanomedicines: some case studies. Nanomedicine 13, 539–554.
Borchert, H., Shevchenko, E.V., Robert, A., Mekis, I., Kornowski, A., Grübel, G., Weller, Gómez, G., Pikal, M.J., Rodríguez-Hornedo, N., 2001. Effect of initial buffer composition
H., 2005. Determination of nanocrystal sizes: a comparison of TEM, SAXS, and XRD on pH changes during far-from-equilibrium freezing of sodium phosphate buffer so-
studies of highly monodisperse CoPt3 particles. Langmuir 21, 1931–1936. lutions. Pharm Res 18, 90–97.
Bracq, E., Lahiani-Skiba, M., Guerbet, M., 2008. Ethical Observations on the Choice of Grau, M., Müller, R., 1998. Increase of dissolution velocity and solubility of poorly soluble
Parenteral Solvents: Choice of Parenteral Solvent…. Drug Dev Ind Pharm 34, drugs by formulation as nanosuspension, Proceedings 2nd World Meeting APGI/APV.
1306–1310. Paris 623–624.
Butler, J.M., Dressman, J.B., 2010. The developability classification system: application of Higgins, J.P., Arrivo, S.M., Thurau, G., Green, R.L., Bowen, W., Lange, A., Templeton,
biopharmaceutics concepts to formulation development. Journal of pharmaceutical A.C., Thomas, D.L., Reed, R.A., 2003. Spectroscopic approach for on-line monitoring
sciences 99, 4940–4954. of particle size during the processing of pharmaceutical nanoparticles. Anal. Chem.
Caputo, F., Clogston, J., Calzolai, L., Rösslein, M., Prina-Mello, A., 2019. Measuring 75, 1777–1785.
particle size distribution of nanoparticle enabled medicinal products, the joint view Innings, F., Hultman, E., Forsberg, F., Prakash, B., 2011. Understanding and analysis of
of EUNCL and NCI-NCL. A step by step approach combining orthogonal measure- wear in homogenizers for processing liquid food. Wear 271, 2588–2598.
ments with increasing complexity. J. Control. Release. Jacobs, C., Kayser, O., Müller, R., 2000. Nanosuspensions as a new approach for the
Chan, H.-K., Kwok, P.C.L., 2011. Production methods for nanodrug particles using the formulation for the poorly soluble drug tarazepide. Int. J. Pharm. 196, 161–164.
bottom-up approach. Adv. Drug Deliv. Rev. 63, 406–416. Jacobs, C., Müller, R.H., 2002. Production and characterization of a budesonide nano-
Chapter85, 2007. Bacterial Endotoxins test, The United States Pharmacopeia, The suspension for pulmonary administration. Pharm. Res. 19, 189–194.
National Formulary, USP30/ NF25. The United States Pharmacopeial Convention, Karan, S.M., Lojeski, E.W., Haynes, D.H., Bina, S., Wesche, D.L., Boedeker, B.H., Muldoon,
Rockville, pp. 109-113. S.M., 1996. Intravenous lecithin-coated microcrystals of dantrolene are effective in
Chaubal, M.V., Popescu, C., 2008. Conversion of nanosuspensions into dry powders by the treatment of malignant hyperthermia: an investigation in rats, dogs, and swine.
spray drying: a case study. Pharm res 25, 2302–2308. Anesthesia & Analgesia 82, 796–802.
Chen, M.-L., John, M., Lee, S.L., Tyner, K.M., 2017. Development considerations for na- Keck, C.M., Müller, R.H., 2006. Drug nanocrystals of poorly soluble drugs produced by
nocrystal drug products. The AAPS journal 19, 642–651. high pressure homogenisation. Eur. J. Pharm. Biopharm. 62, 3–16.
Chen, W., Zuo, H., Zhang, E., Li, L., Henrich-Noack, P., Cooper, H.M., Qian, Y., Xu, Z.P., Keck, C.M., Müller, R.H., 2008. Size analysis of submicron particles by laser dif-
2018. Brain targeting delivery facilitated by ligand-functionalized layered double fractometry—90% of the published measurements are false. Int. J. Pharm. 355,
hydroxide nanoparticles. ACS Appl Mater Interfaces. 150–163.
Chen, X., Vaughn, J.M., Yacaman, M.J., Williams III, R.O., Johnston, K.P., 2004. Rapid Kesisoglou, F., Mitra, A., 2012. Crystalline nanosuspensions as potential toxicology and
dissolution of high-potency danazol particles produced by evaporative precipitation clinical oral formulations for BCS II/IV compounds. The AAPS journal 14, 677–687.
into aqueous solution. J. Pharm. Sci. 93, 1867–1878. Kesisoglou, F., Panmai, S., Wu, Y., 2007. Nanosizing—oral formulation development and
Cho, C.-F., Ablack, A., Leong, H.-S., Zijlstra, A., Lewis, J., 2011. Evaluation of nano- biopharmaceutical evaluation. Adv. Drug Deliv. Rev. 59, 631–644.
particle uptake in tumors in real time using intravital imaging. J VIS EXP, e2808. Kim, S., Lee, J., 2011. Folate-targeted drug-delivery systems prepared by nano-commi-
Crist, R.M., Grossman, J.H., Patri, A.K., Stern, S.T., Dobrovolskaia, M.A., Adiseshaiah, nution. Drug Dev Ind Pharm 37, 131–138.
P.P., Clogston, J.D., McNeil, S.E., 2013. Common pitfalls in nanotechnology: lessons Kipp, J., 2004. The role of solid nanoparticle technology in the parenteral delivery of
learned from NCI's Nanotechnology Characterization Laboratory. Integr. Biol. 5, poorly water-soluble drugs. Int. J. Pharm. 284, 109–122.
66–73. Knapp, J.Z., Barber, T.A., Lieberman, A., 1996. Liquid-and surface-borne particle mea-
Dave, R., Patel, R., Patel, J., Chauhan, H., 2012. Effect of cryoprotectant on lyophilisation surement handbook. Marcel Dekker, New York.
of doxorubicin—HCl loaded chitosan nanoparticles. Int J Pharm Life Sci 3, Krause, K.P., Kayser, O., Mäder, K., Gust, R., Müller, R., 2000. Heavy metal contamination
1769–1772. of nanosuspensions produced by high-pressure homogenisation. Int. J. Pharm. 196,
De Waard, H., Hinrichs, W., Frijlink, H., 2008. A novel bottom–up process to produce 169–172.
drug nanocrystals: controlled crystallization during freeze-drying. J. Control. Release Kwade, A., 1999. Wet comminution in stirred media mills—research and its practical
128, 179–183. application. Powder Technol. 105, 14–20.
Dempah, K.E., Lubach, J.W., Munson, E.J., 2017. Characterization of the particle size and Lakshmi, P., Kumar, G.A., 2010. Nanosuspension technology: A review. Int J Pharm Sci 2,
polydispersity of dicumarol using solid-state NMR spectroscopy. Mol. Pharm. 14, 35–40.
856–865. Leong, H.S., Butler, K.S., Brinker, C.J., Azzawi, M., Conlan, S., Dufès, C., Owen, A.,

13
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

Rannard, S., Scott, C., Chen, C., 2019. On the issue of transparency and reproduci- efficacy through altered pharmacokinetics in the rat. Int. J. Pharm. 339, 251–260.
bility in nanomedicine. Nat. Nanotechnol. 14, 629–635. Rabinow, B.E., 2004. Nanosuspensions in drug delivery. Nat. Rev. Drug Discov 3, 785.
Leong, H.S., Steinmetz, N.F., Ablack, A., Destito, G., Zijlstra, A., Stuhlmann, H., Reid, B.D., 1995. Gamma processing technology: an alternative technology for terminal
Manchester, M., Lewis, J.D., 2010. Intravital imaging of embryonic and tumor neo- sterilization of parenterals. PDA J. Pharm. Sci. Technol. 49, 83–89.
vasculature using viral nanoparticles. Nat. Protoc. 5, 1406. Roethlisberger, D., Mahler, H.-C., Altenburger, U., Pappenberger, A., 2017. If Euhydric
Leung, D.H., Lamberto, D.J., Liu, L., Kwong, E., Nelson, T., Rhodes, T., Bak, A., 2014. A and Isotonic Do Not Work, What Are Acceptable pH and Osmolality for Parenteral
new and improved method for the preparation of drug nanosuspension formulations Drug Dosage Forms? J. Pharm. Sci. 106, 446–456.
using acoustic mixing technology. Int. J. Pharm. 473, 10–19. Salazar, J., Müller, R.H., Möschwitzer, J.P., 2014. Combinative particle size reduction
Li, M., Alvarez, P., Orbe, P., Bilgili, E., 2018. Multi-faceted Characterization of Wet-milled technologies for the production of drug nanocrystals. Journal of pharmaceutics 2014.
Griseofulvin Nanosuspensions for Elucidation of Aggregation State and Stabilization Scherrer, P., 1918. Bestimmung der Groˇsse und der inneren Struktur von Kolloidteilchen
Mechanisms. AAPS PharmSciTech 19, 1789–1801. mittels Roˇntgenstrahlen. Nachrichten von der Gesellschaft der Wissenschaften,
Li, M., Yaragudi, N., Afolabi, A., Dave, R., Bilgili, E., 2015. Sub-100 nm drug particle Goˇttingen, pp. 98–100.
suspensions prepared via wet milling with low bead contamination through novel Shalaev, E.Y., Johnson-Elton, T.D., Chang, L., Pikal, M.J., 2002. Thermophysical prop-
process intensification. Chem Eng Sci. 130, 207–220. erties of pharmaceutically compatible buffers at sub-zero temperatures: implications
Li, P., Zhao, L., 2007. Developing early formulations: practice and perspective. Int. J. for freeze-drying. Pharm Res 19, 195–201.
Pharm. 341, 1–19. Sharma, S., Singh, J., Verma, A., Teja, B.V., Shukla, R.P., Singh, S.K., Sharma, V., Konwar,
Li, W., Yang, Y., Tian, Y., Xu, X., Chen, Y., Mu, L., Zhang, Y., Fang, L., 2011. Preparation R., Mishra, P., 2016. Hyaluronic acid anchored paclitaxel nanocrystals improves
and in vitro/in vivo evaluation of revaprazan hydrochloride nanosuspension. Int. J. chemotherapeutic efficacy and inhibits lung metastasis in tumor-bearing rat model.
Pharm. 408, 157–162. RSC Adv. 6, 73083–73095.
Liu, T., Müller, R.H., Möschwitzer, J.P., 2018. Production of drug nanosuspensions: effect Shegokar, R., Müller, R.H., 2010. Nanocrystals: industrially feasible multifunctional for-
of drug physical properties on nanosizing efficiency. Drug Dev Ind Pharm. 44, mulation technology for poorly soluble actives. Int. J. Pharm. 399, 129–139.
233–242. Shegokar, R., Singh, K.K., 2011. Surface modified nevirapine nanosuspensions for viral
Liu, Y., Huang, L., Liu, F., 2010. Paclitaxel nanocrystals for overcoming multidrug re- reservoir targeting: In vitro and in vivo evaluation. Int. J. Pharm. 421, 341–352.
sistance in cancer. Mol. Pharm. 7, 863–869. Shegokar, R., Singh, K.K., 2012. Nevirapine nanosuspensions: stability, plasma compat-
Lück, M., Paulke, B.R., Schröder, W., Blunk, T., Müller, R., 1998. Analysis of plasma ibility and sterilization. J Pharm Investig. 42, 257–269.
protein adsorption on polymeric nanoparticles with different surface characteristics. Sheokand, S., Reddy, V., Bansal, A.K., 2018. Pharmaceutical Nanocrystals: From
J. Biomed. Mater. Res. 39, 478–485. Fundamentals to Advances. Pharma Times 50, 20–25.
Maeda, H., 2015. Toward a full understanding of the EPR effect in primary and metastatic Shi, Y., Porter, W., Merdan, T., Li, L.C., 2009. Recent advances in intravenous delivery of
tumors as well as issues related to its heterogeneity. Adv. Drug Deliv. Rev. 91, 3–6. poorly water-soluble compounds. Expert Opin. Drug Deliv. 6, 1261–1282.
Merisko-Liversidge, E., Liversidge, G.G., 2011. Nanosizing for oral and parenteral drug Shubar, H.M., Dunay, I.R., Lachenmaier, S., Dathe, M., Bushrab, F.N., Mauludin, R.,
delivery: a perspective on formulating poorly-water soluble compounds using wet Müller, R.H., Fitzner, R., Borner, K., Liesenfeld, O., 2009. The role of apolipoprotein E
media milling technology. Adv. Drug Deliv. Rev. 63, 427–440. in uptake of atovaquone into the brain in murine acute and reactivated tox-
Merisko-Liversidge, E., Sarpotdar, P., Bruno, J., Hajj, S., Wei, L., Peltier, N., Rake, J., oplasmosis. J Drug Target 17, 257–267.
Shaw, J., Pugh, S., Polin, L., 1996. Formulation and antitumor activity evaluation of Sigfridsson, K., Forssén, S., Holländer, P., Skantze, U., de Verdier, J., 2007. A formulation
nanocrystalline suspensions of poorly soluble anticancer drugs. Pharm. Res. 13, comparison, using a solution and different nanosuspensions of a poorly soluble
272–278. compound. Eur J Pharm Biopharm. 67, 540–547.
Moller, N., Jensen, V., 1970. Studies on particle size problems. V. Surface area stabilities Sinha, B., Müller, R.H., Möschwitzer, J.P., 2013. Bottom-up approaches for preparing
of micronized powders in relation to humidity and heat sterilization. Dansk Tidsskr. drug nanocrystals: formulations and factors affecting particle size. Int. J. Pharm. 453,
Farm. 44, 301–310. 126–141.
Möschwitzer, J.P., 2013. Drug nanocrystals in the commercial pharmaceutical develop- Sohn, J.S., Yoon, D.-S., Sohn, J.Y., Park, J.-S., Choi, J.-S., 2017. Development and eva-
ment process. Int. J. Pharm. 453, 142–156. luation of targeting ligands surface modified paclitaxel nanocrystals. Mater. Sci. Eng.
Mouton, J., Van Peer, A., De Beule, K., Van Vliet, A., Donnelly, J., Soons, P., 2006. C. 72, 228–237.
Pharmacokinetics of itraconazole and hydroxyitraconazole in healthy subjects after Spreen, W., Williams, P., Margolis, D., Ford, S.L., Crauwels, H., Lou, Y., Gould, E.,
single and multiple doses of a novel formulation. Antimicrob. Agents Chemother 50, Stevens, M., Piscitelli, S., 2014. Pharmacokinetics, safety, and tolerability with repeat
4096–4102. doses of GSK1265744 and rilpivirine (TMC278) long-acting nanosuspensions in
Müller-Goymann, C., 2004. Physicochemical characterization of colloidal drug delivery healthy adults. JAIDS-J ACQ IMM DEF 67, 487–492.
systems such as reverse micelles, vesicles, liquid crystals and nanoparticles for topical Starostina, N., West, P., 2006. Part II: sample preparation for AFM particle character-
administration. Eur J Pharm Biopharm. 58, 343–356. ization. Pacific Nanotechnology Inc., Santa Clara, CA, pp. 1–10.
Müller, R., Jacobs, C., Kayser, O., 2001. Nanosuspensions as particulate drug formulations Suri, G.S., Kaur, A., Sen, T., 2016. A recent trend of drug-nanoparticles in suspension for
in therapy: rationale for development and what we can expect for the future. Adv. the application in drug delivery. Nanomedicine 11, 2861–2876.
Drug Deliv. Rev. 47, 3–19. Swierczewska, M., Crist, R.M., McNeil, S.E., 2018. Evaluating nanomedicines: obstacles
Müller, R.H., Benita, S., Böhm, B.H., 1998. Emulsions and nanosuspensions for the for- and advancements, Characterization of Nanoparticles Intended for Drug Delivery.
mulation of poorly soluble drugs. Medpharm Scientific Publishers, Stuttgart. Springer 3–16.
Müller, R.H., Gohla, S., Keck, C.M., 2011. State of the art of nanocrystals–special features, Teeranachaideekul, V., Junyaprasert, V.B., Souto, E.B., Müller, R.H., 2008. Development
production, nanotoxicology aspects and intracellular delivery. Eur J Pharm Biopharm of ascorbyl palmitate nanocrystals applying the nanosuspension technology. Int. J.
78, 1–9. Pharm. 354, 227–234.
Müller, R.H., Hildebrand, G.E., 1996. Zetapotential und Partikelladung in der Laborpraxis ten Tije, A.J., Verweij, J., Loos, W.J., Sparreboom, A., 2003. Pharmacological effects of
(Einführung in die Theorie praktische Messdurchführung Dateninterpretation). formulation vehicles: implications for cancer chemotherapy. Clin. Pharmacokinet.
Paperback APV. 42, 665–685.
Müller, R.H., Peters, K., 1998. Nanosuspensions for the formulation of poorly soluble Thakkar, S., Sharma, D., Misra, M., 2018. Comparative evaluation of electrospraying and
drugs: I. Preparation by a size-reduction technique. Int J Pharm 160, 229–237. lyophilization techniques on solid state properties of Erlotinib nanocrystals:
Niwa, T., Danjo, K., 2013. Design of self-dispersible dry nanosuspension through wet Assessment of In-vitro cytotoxicity. Eur. J. Pharm. Sci. 111, 257–269.
milling and spray freeze-drying for poorly water-soluble drugs. Eur. J. Pharm. Sci. 50, Townson, J.L., Lin, Y.-S., Agola, J.O., Carnes, E.C., Leong, H.S., Lewis, J.D., Haynes, C.L.,
272–281. Brinker, C.J., 2013. Re-examining the size/charge paradigm: differing in vivo char-
Noh, J.-K., Naeem, M., Cao, J., Lee, E.H., Kim, M.-S., Jung, Y., Yoo, J.-W., 2016. acteristics of size-and charge-matched mesoporous silica nanoparticles. J. Am. Chem.
Herceptin-functionalized pure paclitaxel nanocrystals for enhanced delivery to HER2- Soc. 135, 16030–16033.
postive breast cancer cells. Int. J. Pharm. 513, 543–553. Van Eerdenbrugh, B., Vermant, J., Martens, J.A., Froyen, L., Van Humbeeck, J.,
Pace, S.N., Pace, G.W., Parikh, I., Mishra, A.K., 1999. Novel injectable formulations of Augustijns, P., Van den Mooter, G., 2009. A screening study of surface stabilization
insoluble drugs. Pharm Technol 23, 116–134. during the production of drug nanocrystals. J. Pharm. Sci. 98, 2091–2103.
Pandey, G., Mittapelly, N., Banala, V.T., Mishra, P.R., 2018. Multifunctional Verma, S., Huey, B.D., Burgess, D.J., 2009a. Scanning probe microscopy method for na-
Glycoconjugate Assisted Nanocrystalline Drug Delivery for Tumor Targeting and nosuspension stabilizer selection. Langmuir 25, 12481–12487.
Permeabilization of Lysosomal-Mitochondrial Membrane. ACS applied materials & Verma, S., Kumar, S., Gokhale, R., Burgess, D.J., 2011. Physical stability of nanosus-
interfaces 10, 16964–16976. pensions: investigation of the role of stabilizers on Ostwald ripening. Int. J. Pharm.
Patravale, V., Date, A.A., Kulkarni, R., 2004. Nanosuspensions: a promising drug delivery 406, 145–152.
strategy. J. Pharm. Pharmacol. 56, 827–840. Verma, S., Lan, Y., Gokhale, R., Burgess, D.J., 2009b. Quality by design approach to
Peltonen, L., 2018. Design space and QbD approach for production of drug nanocrystals understand the process of nanosuspension preparation. Int. J. Pharm. 377, 185–198.
by wet media milling techniques. Pharmaceutics 10, 104. Wahlstrom, J.L., Chiang, P.-C., Ghosh, S., Warren, C.J., Wene, S.P., Albin, L.A., Smith,
Peters, K., Leitzke, S., Diederichs, J., Borner, K., Hahn, H., Müller, R., Ehlers, S., 2000. M.E., Roberds, S.L., 2007. Pharmacokinetic evaluation of a 1, 3-dicyclohexylurea
Preparation of a clofazimine nanosuspension for intravenous use and evaluation of its nanosuspension formulation to support early efficacy assessment. Nanoscale Res Lett
therapeutic efficacy in murine Mycobacterium avium infection. J Antimicrob 2, 291.
Chemother. 45, 77–83. Wallis, K., Müller, R., 1993. Determination of the surface hydrophobicity of colloidal
Polomska, A., Gauthier, M.A., Leroux, J.C., 2017. In Vitro and In Vivo Evaluation of dispersions by mini-hydrophobic interaction chromatography. Pharm. Ind. 55,
PEGylated Layer-by-Layer Polyelectrolyte-Coated Paclitaxel Nanocrystals. Small 13, 1124–1128.
1602066. Wang, W., 2015. Tolerability of hypertonic injectables. Int. J. Pharm. 490, 308–315.
Rabinow, B., Kipp, J., Papadopoulos, P., Wong, J., Glosson, J., Gass, J., Sun, C.-S., Wang, Z., Chen, J.-F., Le, Y., Shen, Z.-G., Yun, J., 2007. Preparation of ultrafine beclo-
Wielgos, T., White, R., Cook, C., 2007. Itraconazole IV nanosuspension enhances methasone dipropionate drug powder by antisolvent precipitation. Ind. Eng. Chem.

14
D. Patel, et al. International Journal of Pharmaceutics 586 (2020) 119555

Res. 46, 4839–4845. 338–343.


Wanning, S., Süverkrüp, R., Lamprecht, A., 2015. Pharmaceutical spray freeze drying. Int. Xu, L.-M., Zhang, Q.-X., Zhou, Y., Zhao, H., Wang, J.-X., Chen, J.-F., 2012. Engineering
J. Pharm. 488, 136–153. drug ultrafine particles of beclomethasone dipropionate for dry powder inhalation.
Willems, L., Van der Geest, R., De Beule, K., 2001. Itraconazole oral solution and in- Int. J. Pharm. 436, 1–9.
travenous formulations: a review of pharmacokinetics and pharmacodynamics. J. Yalkowsky, S.H., 1999. Solubility and solubilization in aqueous media. American
Clin. Pharm. Ther. 26, 159–169. Chemical Society Washington, DC.
Williamson, G., Hall, W., 1953. X-ray line broadening from filed aluminium and wolfram. Young, R.A., 1993. The Rietveld Method. International union of crystallography.
Acta metallurgica 1, 22–31. Yuan, Q., Wang, Y., Song, R., Hou, X., Yu, K., Zheng, J., Zhang, J., Han, J., Pu, X., Zong,
Wong, J., Brugger, A., Khare, A., Chaubal, M., Papadopoulos, P., Rabinow, B., Kipp, J., L., 2019. Study on Formulation, in vivo Exposure and Passive Targeting of
Ning, J., 2008. Suspensions for intravenous (IV) injection: a review of development, Intravenous Itraconazole Nanosuspensions. Front Pharmacol 10, 225.
preclinical and clinical aspects. Adv. Drug Deliv. Rev. 60, 939–954. Zhang, H., Hollis, C.P., Zhang, Q., Li, T., 2011. Preparation and antitumor study of
Wu, L., Zhang, J., Watanabe, W., 2011. Physical and chemical stability of drug nano- camptothecin nanocrystals. Int. J. Pharm. 415, 293–300.
particles. Adv. Drug Deliv. Rev. 63, 456–469. Zheng, J.Y., Bosch, H.W., 1997. Sterile filtration of NanoCrystal™ drug formulations. Drug
Xiong, R., Lu, W., Li, J., Wang, P., Xu, R., Chen, T., 2008. Preparation and character- Dev. Ind. Pharm. 23, 1087–1093.
ization of intravenously injectable nimodipine nanosuspension. Int. J. Pharm. 350,

15

You might also like