You are on page 1of 20

International Journal of Pharmaceutics 453 (2013) 233–252

Contents lists available at SciVerse ScienceDirect

International Journal of Pharmaceutics


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

Review

Melt extrusion with poorly soluble drugs


Sejal Shah, Sindhuri Maddineni, Jiannan Lu, Michael A. Repka ∗
Department of Pharmaceutics, Pii Center for Pharmaceutical Technology, School of Pharmacy, The University of Mississippi, University, MS 38677-1848, United States

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

Article history: Melt extrusion (ME) over recent years has found widespread application as a viable drug delivery option in
Received 7 June 2012 the drug development process. ME applications include taste masking, solid-state stability enhancement,
Received in revised form 31 October 2012 sustained drug release and solubility enhancement. While ME can result in amorphous or crystalline solid
Accepted 1 November 2012
dispersions depending upon several factors, solubility enhancement applications are centered around
Available online 20 November 2012
generating amorphous dispersions, primarily because of the free energy benefits they offer. In line with
the purview of the current issue, this review assesses the utility of ME as a means of enhancing solubil-
Keywords:
ity of poorly soluble drugs/chemicals. The review describes major processing aspects of ME technology,
Melt extrusion
Poorly water soluble drugs
definition and understanding of the amorphous state, manufacturability, analytical characterization and
Dissolution rate enhancement biopharmaceutical performance testing to better understand the strength and weakness of this formu-
Solid dispersion lation strategy for poorly soluble drugs. In addition, this paper highlights the potential advantages of
Amorphous system employing a fusion of techniques, including pharmaceutical co-crystals and spray drying/solvent evapo-
Solid solution ration, facilitating the design of formulations of API exhibiting specific physico-chemical characteristics.
Thermoplastic polymers Finally, the review presents some successful case studies of commercialized ME based products.
© 2012 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 234
2. Solubilization using melt extrusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 235
3. Manufacturing aspects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
3.1. Equipment for melt extrusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
3.1.1. Downstream processing equipment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
3.2. Melt extrusion processing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 237
3.3. Screw design and configuration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
3.4. Process analytical technology (PAT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
3.5. Processing parameters and scale-up . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 238
3.6. Screening criteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239
4. Solubility enhancement using hydrophilic polymers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 239
5. Characterization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
5.1. Spectroscopic techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
5.1.1. X-ray diffraction methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 242
5.1.2. IR and Raman spectroscopy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 243
5.2. Thermal techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 244
5.2.1. Differential scanning calorimetry . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 244
5.2.2. Isothermal microcalorimetry (IC) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245
5.3. Microscopic techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245
5.4. Microthermal analysis (Micro-TA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245
5.5. Water vapor sorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245
6. Innovation in melt extrusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
6.1. Formation of co-crystals using melt extrusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
6.2. Fusion of solvent evaporation/spray drying techniques with melt extrusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247

∗ Corresponding author. Tel.: +1 662 915 1155; fax: +1 662 915 1177.
E-mail address: marepka@olemiss.edu (M.A. Repka).

0378-5173/$ – see front matter © 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.ijpharm.2012.11.001
234 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

7. Marketed formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247


8. Comparison of different solid dispersion techniques . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
9. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249

1. Introduction Crystalline solid dispersions are generally designed to achieve


controlled drug release profiles for highly soluble drugs (Dierickx
The application of molecular modeling and high throughput et al., 2012; Ma et al., 2012; Roblegg et al., 2011; Verhoeven et al.,
drug innovation has led to discovery of numerous new chemi- 2009).
cal entities as potential therapeutic agents. However, obstacles Micro or nano crystalline solid dispersions also find applica-
associated with solubility, bioavailability and toxicity undermine tion in solubility enhancement. Thommes et al. (2011) describe a
their development and commercialization. The enhancement of system termed as solid crystal suspension (crystalline drug sus-
oral bioavailability of poorly water-soluble drugs remains one pended in crystalline carrier matrix) analogous to the crystalline
of the most challenging aspects of the drug development pro- solid dispersion, prepared using top down technique. This solid
cess. Pharmaceutical scientists, over the years have successfully crystal suspension was designed to improve the dissolution of three
developed several strategies for design of delivery systems for poorly water-soluble drugs. The system comprised of high melting
poorly soluble drugs. Solid dispersions prepared by melt extru- point drugs like griseofulvin (10% and 50%), spironolactone (10%)
sion (ME) technology is one such strategy that over the past and phenytoin (10%) and mannitol as a rapidly recrystallizing car-
three decades has resulted in several unique drug delivery rier phase. X-ray diffraction profile revealed the crystalline nature
systems. of extruded formulation similar to the combined pattern of crys-
Melt extrusion has been successfully applied to enhance solu- talline drug and excipients respectively. The dissolution rates of
bility of poorly soluble drugs (Feng et al., 2011; Lakshman et al., the extrudates were considerably higher than that for the pure
2008; Liu et al., 2012b; Sakurai et al., 2012; Tho et al., 2010). Sol- drug and physical mixture, attributable to the enhanced wetting
ubility enhancement in the melt extrusion process occurs through of the API by mannitol. Moreover, the crystalline character of the
dispersion of a poorly soluble drug(s) in a polymeric (or lipid) formulation was devoid of any physical stability issues, which is an
carrier matrix essentially forming a solid dispersion. According to inherent limitation with amorphous formulations. In addition, the
Chiou and Riegelman a solid dispersion is defined as ‘dispersion of melt extrusion processing reduced the particle size of API resulting
one or more API in an inert carrier in the solid state, prepared by in uniform distribution of API in the rapidly crystallizing manni-
either melting, solvent or the combined melting-solvent method’. tol matrix. Hence, the combined effect of melt processing and the
Further, they classify solid dispersions as eutectic mixtures, solid solid suspension of crystalline API in hydrophilic crystalline matrix
solutions and glass solutions based on the physical state of the drug appear to be a reasonable approach suitable for drugs that are dif-
and the carrier (Chiou and Riegelman, 1971). During melt extru- ficult to stabilize in the amorphous form.
sion of a drug within a polymer binary mixture, the drug could The bottom-up manufacturing process using hot melt extrusion
remain molecularly dispersed within the polymer or exists as an is described in a recent invention (Chatterji et al., 2012) that illus-
amorphous or crystalline phase. Hence, based on the molecular trates the formation of controlled crystalline solid dispersion of
state of the API distributed in the carrier phase and characteristic API from its super cooled liquid state. The invention describes a
thermal properties (drug melting temperature Tm /glass transition process, wherein the crystalline API is converted to non-crystalline
temperature Tg or melting temperature Tmc of the carrier phase form by application of heat and shear up to 1/4th to 3/4th of the
as identified by DSC) of the system, solid dispersions prepared by barrel length followed by a recrystallization zone in the remaining
HME can be categorized as crystalline solid dispersions, amorphous barrel length, wherein cooling is applied. The cooling of the bar-
solid dispersions, and solid solutions (also regarded as a subgroup rel initiated API nucleation that promoted crystal growth while
of amorphous solid dispersions) (Fig. 1) (Janssens and Van den the shearing action of the screws evenly distributes the nuclei and
Mooter, 2009; Leuner and Dressman, 2000; Serajuddin, 1999). A hence controls the mean particle diameter of the newly formed
thorough understanding of structure of a solid dispersion, partic- crystalline API. The particle size of newly formed crystalline API
ularly the existing physical form of a drug in the carrier matrix is is significantly lesser than the bulk API. Recrystallization of the
required to predict the stability, solubility and hence bioavailability API is controlled by carrier formulation design and ME process
of melt extrudates. parameters like barrel temperature, feed rate, etc. The crystalline
Crystalline solid dispersions are systems wherein the crystalline drug, dalcetrapib, is unstable in its amorphous state; hence, the
drug substance is dispersed into an amorphous carrier matrix. The aforementioned processing technique was found to be a rather
DSC profile for such a system is characterized by the presence of a suitable method of production. In addition, the rapid dissolution
melting endotherm (Tm ) corresponding to the crystalline API and a was observed as compared to its micronized form. However, this
characteristic glass transition temperature (Tg ) corresponding to approach faces technical challenges such as maintaining consis-
the amorphous carrier. Crystalline solid dispersions can be pre- tent batch-to-batch crystallization (particle size of crystals), and
pared using either the top-down or bottom-up techniques. The detection of residual amorphous drug content.
bottom-up process, encompassing the growth of small particles Amorphous solid dispersions result when melt extruded
from individual molecules is attributed to the immiscibility or drug–polymer is cooled at a rate that does not allow the drug to
supersaturation of API in the carrier phase either in molten or solid recrystallize or processed at temperatures where drug melts but
(after cooling) form of the extrudates. It is important to note that, remains immiscible with the carrier. Such processing results in
in either of the techniques for the drug to be 100% crystalline in the kinetic entrapment of the drug in its amorphous state. Although,
carrier matrix, it should be completely immiscible with the carrier these types of systems exhibit increased rate of dissolution due
phase at the employed processing conditions. Moreover, for the top to high thermodynamic activity, they have a potential to revert to
down technique, the Tm of the drug is essentially greater than the the more stable crystalline form (Janssens and Van den Mooter,
Tg of amorphous carrier. 2009; Leuner and Dressman, 2000). In a solid solution, the drug
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 235

Fig. 1. States of solid dispersion.

molecule is molecularly dissolved in the polymeric carrier matrix While HME has several advantages, it is limited because of
and exhibits a single glass transition temperature (Tg ). An amor- its inability to process heat sensitive or high melting point
phous solid solution is a pharmaceutically desirable single-phase drugs. Moreover, HME is an easily scalable method for large-
system preferably including an amorphous polymer as the carrier, scale commercial manufacture but HME (primarily the equipment)
a drug in its high-energy state, as well as other excipients such as is generally not amenable for scale-down manufacturing and
processing aids, recrystallization inhibitors, and wetting agents. screening (processing mg to g quantities of drug product) typi-
Melt extrusion application for solubility enhancement aims at cally required in early stage development. In addition, the analytical
generating amorphous solid dispersions. Amorphous solid dis- characterization methods are limited in terms of detecting phase
persions not only offer the inherent free energy benefits of an separation in amorphous dispersions or quantifying trace levels of
amorphous system, but also provide maximum specific surface area crystallinity. The strategies adopted to overcome these limitations
and higher saturation solubility, which ultimately increase drug (miniaturized screening methods for early stage development, local
solubility. Thorough understanding of the physicochemical proper- and global analytical methods for detection of phase separation,
ties of amorphous solid dispersions and their corresponding in vivo quantification methods for residual crystallinity) are discussed in
behavior is required for the realization of their true potential in the sufficient detail in subsequent sections.
pharmaceutical industry. Van den Mooter has extensively reviewed This review highlights major aspects of melt extrusion
the application of amorphous solid dispersions as an interesting technology, the amorphous state, manufacturing, analytical char-
strategy to increase the bioavailability of poorly soluble drugs (Van acterization and biopharmaceutical testing to better understand
den Mooter, 2012). Van den Mooter discusses important aspects the strengths and weaknesses of this formulation strategy for
such as the amorphous state, manufacturing, characterization, and poorly soluble drugs. Furthermore, this paper presents some suc-
biopharmaceutical testing for better understanding of the strength cessful case studies of commercialized ME based products.
and weakness of solid dispersions.
It is a general understanding that amorphous solid dispersions
enhance the solubility and hence improves bioavailability of drugs; 2. Solubilization using melt extrusion
however, it is certainly not a universal rule applicable to all drugs.
Newman et al. make a comparative assessment of the performance Successful solubilization in melt extrusion is determined by
of amorphous solid dispersions, described in 40 research papers several factors, which are material (drug and carrier proper-
(Newman et al., 2012). The results from this comparative assess- ties), process (processing temperature, shear, etc.) and equipment
ment are classified into 3 categories (i) amorphous dispersions with (design and operating conditions) related. We discuss the mate-
improved bioavailability (82% of the cases); (ii) amorphous disper- rial and process related factors in this section, equipment related
sion with no change or similar bioavailability to reference material factors are discussed in the subsequent section. Processing temper-
(10% of the cases); (iii) amorphous dispersions with lower bioavail- ature is an important parameter in melt extrusion. Ideal processing
ability (8% of the cases). An analysis of these studies revealed several temperature is selected based on the melting temperature (Tm ) of
in vitro and in vivo variables that could influence the results. Based the drug, Tmc /Tg of the carrier and is generally greater than the
on these variables, they make several recommendations for consid- Tmc /Tg of the carrier. Such high processing temperatures ensure
erations in the standardized performance evaluation of amorphous that the carrier is softened (often melted) and possess low viscosity,
solid dispersions. thereby permitting extrusion. Processing aids such as plasticizers
236 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

can be added to improve the flow of the melt. Ideally, the Tmc or Tg rate of the micronized particles was improved due to particle deag-
of carrier should not be too high since high temperature processing gregation and enhanced wetting (Miller et al., 2007).
may potentially lead to drug degradation. Like process temperature, the polymeric carrier is also impor-
DiNunzio et al. (2012) classify melt extrusion processing into tant and has a significant impact on the success rate of the solid
two distinct regimes, based on extrusion temperature (Tprocess ) dispersions. Inherent melt solubility is an important aspect for car-
employed and the melting temperature (Tm ) of the drug. In the sol- rier selection, as this significantly limits the range of materials
ubilization regime, Tprocess < Tm whereas in the miscibility regime, that can be used as carriers for solubilization. The carrier should
Tprocess ≥ Tm however the glass transition temperature (Tg ) of the be heat stable at the employed processing temperature, pharma-
carrier is well above the Tprocess in both the regimes (DiNunzio et al., cologically inert and safe. In addition, thermal stability and melt
2012). viscosity are important considerations for melt extrusion carri-
The miscibility of drug phase in the molten carrier matrix ers. Aqueous solubility of the carrier is an obvious requirement.
predominantly dictates the successful preparation and hence sta- Janssens and Van den Mooter (2009) have extensively reviewed
bilization of amorphous solid dispersion in the miscibility regime. the most important carrier properties and provide an overview of
In addition, the appropriate selection of screw configuration and the polymers that have been used in commercial marketed for-
the shearing action of screws during hot melt processing assist in mulations. Physical stability of amorphous solid dispersion could
homogeneous dispersion of the drug in the molten carrier phase. be improved by anti-plasticization effect of polymers (increasing
Processing under miscibility regime is limited due to its inability to the viscosity of the binary system and decreasing the diffusion of
process high melting point drugs. Various approaches such as dif- drug molecules) that would raise the glass transition temperature
ferential solubility parameter (<7 MPa1/2 ) (Greenhalgh et al., 1999), of the system (Kakumanu and Bansal, 2002; Sathigari et al., 2012;
Floury–Huggins mixing theory (Abu-Diak et al., 2012; Yang et al., Van den Mooter et al., 2001). Hydrogen bonding and hydrophobic
2012; Zhao et al., 2011), Gordon–Taylor equation with appropriate interactions between the drug and polymer are the primary driving
assumptions and modifications (Forster et al., 2001a), determina- forces for the formation of solid dispersions during melt extrusion,
tion of Tg using DSC in conjunction with mid IR and XRPD with inhibition of drug crystallization during subsequent storage of melt
computational analysis (Rumondor et al., 2009a), etc. are employed exudates and achievement and sustainment of supersaturation in
to assess the drug–polymer miscibility. the GI tract.
The processing under solubilization regime is better understood The solubility and miscibility of drug in the polymer are directly
by Noyes–Whitney equation, wherein increased dissolution rates related to the stabilization of an amorphous drug against crystal-
(dM/dt) of crystalline drug in the molten carrier matrix could be lization (Qian et al., 2010). Unfortunately, the amorphous state is
achieved by raising the temperature of the system (i.e. processing less stable than the crystalline state, at times resulting in the recrys-
above the glass transition temperature of carrier phase) that would tallization of drug from solid dispersions during the manufacturing
result in increased equilibrium solubility (C0 ) and diffusivity (D) of process and subsequently during storage (Vasconcelos et al., 2007).
the drug in the molten carrier phase. Below the Tg , which represents a kinetic boundary of molec-
ular mobility, the stability of solid dispersions strongly dependent
dM DA upon kinetics of phase separation and crystallization. Yoshioka et al.
= × (C0 − Ct ) (1)
dt h proposed that at the temperature 50 ◦ C lower than Tg , the molecu-
lar mobility can be neglected and the amorphous solids are stable
Each of the terms in the above equation (Eq. (1)) can be altered to enough over a period of years (Hancock et al., 1995; Yoshioka et al.,
ultimately achieve high dissolution rates (dM/dt), surface area (A) 1995). This concept is popularly referred to as the ‘Tg − 50 ◦ C’ rule.
of the drug can be increased by micronization. Increased dissolu- While this rule is valid in most cases, there are some excep-
tion time (t) can be achieved by increasing the mean residence tions (incompatibility when Tg − Tstorage > 50 ◦ C and stability when
time of the dispersion in the extruder, increased solubility (C0 ) and Tg − Tstorage < 50 ◦ C) (Forster et al., 2001a; Vyazovkin and Dranca,
diffusivity (D) could be achieved by raising the processing tem- 2007). This is probably because the concept takes into account only
perature or by addition of co-solvents, plasticizers, etc. Applying the primary relaxations (“␣” relaxations or global mobility), which
more shears (suitable screw configuration and screw speed) to the are less significant below Tg . Secondary relaxations (“␤” relaxations
system would result in further reducing the viscosity (decrease in or local mobility) therefore also need to be considered as these
boundary layer thickness, h) and thereby enhancing the diffusivity lead to nucleation at temperatures below Tg where the coopera-
and dissolution rate of the drug in the carrier phase. tive “␣” relaxations are negligible (Grzybowska et al., 2010). “␤”
Processing under this regime is most appropriate for high- relaxations are associated with the movement of only a part of
melting point and heat sensitive drugs. a molecule, e.g. polymer side chains and occur on a smaller time
Certain modifications such as micronization of the drug sub- scale than the primary “␣” relaxations (relaxation time being typ-
stance prior to extrusion serve as an effective means to enhance ically < 10−1 s) (Bhattacharya and Suryanarayanan, 2009). Stability
the surface area and hence the drug dissolution rates in the molten even with Tg − Tstorage < 50 ◦ C, is attributed to specific intermolecu-
polymer, thereby improving the melt extrusion processing and the lar interactions (e.g. hydrogen bonding or ion–dipole interactions
performance of melt extruded product. Hughey and co-workers between the drug and polymer) which results in an elevated acti-
describe the preparation of Eudragit® L100-55 based amorphous vation energy for recrystallization (Khougaz and Clas, 2000; Konno
dispersions of micronized Roche Research compound A (ROA). It and Taylor, 2006; Matsumoto and Zografi, 1999; Miyazaki et al.,
was identified that micronization and reduced residence time were 2004; Taylor and Zografi, 1997).
necessary for generating amorphous dispersions. Hughey et al. Presence of moisture (inherent, during processing or stor-
(2010) compare Kinetisol® dispersion technique and the hot melt age) is an important factor resulting in deviation from ideality.
extrusion method as a means to prepare dissolution enhanced solid Moisture due to its plasticizing effect increases the molec-
dispersions of ROA. Miller and co-workers describe the preparation ular mobility and results in depression of the system Tg
of melt extrudates of micronized particles of amorphous itracona- (Marsac et al., 2010; Rumondor et al., 2009b). Moisture induced
zole stabilized with PVP or HPMC. PEO 200 M and poloxamer 407 amorphous–amorphous phase separation has also been studied
were employed as deaggregation and dispersing agents during (Rumondor et al., 2011)
extrusion. Analytical characterization demonstrated that HME did Qian et al. (2010) described challenges in development of solid
not alter properties of micronized particles. Further, dissolution dispersions. In addition, these researchers highlighted investigative
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 237

strategies to better understand the destabilization mechanism of


amorphous solid dispersions.
Successful development of ME based amorphous formulations
depends on a robust preformulation assessment. The selection of
optimal processing conditions depends primarily on the chemical
stability of both the drug and the polymer and the physical proper-
ties of the polymer. Preformulation considerations to establish the
most appropriate process parameters for ME are solubility, drug
melting point (Tm ), glass transition temperature Tg of the polymer,
processing temperature (Tprocess /THME ), drug–polymer miscibility
and melt viscosity of the polymer. These have been extensively
studied by several workers (Breitenbach, 2002; Chokshi et al., 2005;
Forster et al., 2001a,b; Marsac et al., 2006) and have been reviewed
in sufficient detail (Crowley et al., 2007; Repka et al., 2007, 2008)

3. Manufacturing aspects

Based on the ongoing developments in fundamental material Fig. 2. Illustration of a hot melt extrusion film assembly. (Courtesy of Thermo Elec-
tron Corporation, MA.)
sciences and engineering innovations, melt extrusion technology in
the pharmaceutical industry is now considered an evolved science.
The processing equipment for ME has been extensively developed The basic components of an extruder are the feeder, barrel
and it is now a common understanding in the field that formula- (screws or ram), control panel, torque sensors, heating/cooling
tion ultimately defines the equipment design and the processing device, assorted dies, and downstream processing machinery
parameters and vice versa. Furthermore, both formulation and (Fig. 2). Screw extruders and ram extruders are two commonly
processing conditions determine key product quality attributes. used extruders for pharmaceutical processing. Screw extruders can
There are several studies describing these influences during melt be further classified based on certain fundamental properties such
extrusion (Dinunzio et al., 2010a; Repka et al., 1999; Schilling et al., as, number of screws, screw size, the direction of rotation of the
2007). screws and the degree of element intermeshing. Screw extrud-
Early stage formulation development studies should primarily ers used for pharmaceutical applications typically consist of two
focus on the interplay of formulation and processing conditions screws (twin-screw) and are further categorized as co-rotating and
using rational approaches. While the primary aim of early stage counter-rotating, depending on the direction of rotation of the
studies is to develop a basic formulation and a process that supports screw.
late stage development, early stage development also focuses on Co-rotating being used for most applications, with the excep-
the identification of critical process parameters governing product tion that sometimes counter-rotating (non-intermeshing type) is
attributes. Sufficient time and effort is spent in understanding the employed when a small processing window is needed for heat sen-
effect of processing conditions on important process parameters sitive materials. However, the ultimate choice of screw extruder
and hence formulation attributes. depends upon several factors, material properties being the most
Both early and late stage developments employ a Quality by important of them. Unlike a screw extruder, a ram extruder opera-
Design (QbD) based approach wherein target product profile is tes by a positive displacement ram, which can push the material
well defined and critical quality attributes have been established. through the die driven by high pressure.
QbD employs risk assessment by linking material properties and
process parameters to critical quality attributes. Successful under- 3.1.1. Downstream processing equipment
standing of the formulation-process interplay during early stage A number of downstream processing equipment have been
development facilitates the determination of design space and developed and utilized in the hot melt extrusion process meet-
presents an efficient control strategy for critical product quality ing the requirements of final dosage forms. Dies in varying sizes
attributes at late stage development including large-scale man- and shapes are available for formulation development. Flat dies are
ufacturing. Large-scale commercial production requires a robust, utilized for production of films and sheets, circular dies are used
scalable, and transferable process (Food and Drug Administration for pelletization and spheronization whereas the annular dies are
Quality systems approach to pharmaceutical cGMP regulations, used for medical devices and tubing. Air, nitrogen, water, or stain-
2006). For the purpose of this review, the authors emphasize only less steel conveyors are used for the cooling of extrudates. Chill
on the engineering improvisations for stringent process control and rolls have been utilized for film production, wherein adjusting the
hence better melt extruded formulation development. The fun- rotating speed of chill rolls as well as the die opening controls
damental engineering aspects of hot melt extrusion equipment, the thickness of films. Take-up rolls and flakers are also used for
although extremely important, are reviewed in sufficient detail downstream processing of melt-extruded materials.
elsewhere (Breitenbach, 2002; Crowley et al., 2007; Repka et al.,
2012) and hence discussed in limited details herein. 3.2. Melt extrusion processing

3.1. Equipment for melt extrusion In general, the API and polymers are premixed with other excip-
ients or individually fed into the extruder. The physical mixture is
Although developed in the plastic industry, the equipment and further extruded under a defined thermal and mechanical condi-
operational units of melt extruders have been well adapted to tion as a continuous process to generate a final product of uniform
meet the specific demands of pharmaceutical industry and are con- size, shape, and content. From the standpoint of processing, ME can
tinuing to do so. Over the years, engineering and manufacturing be theoretically sub-divided into five steps: (1) feeding, (2) melt-
companies have taken special efforts to design modular pharma- ing and plasticizing, (3) conveying and mixing, (4) venting and (5)
ceutical grade extruders, in accordance with the current Good stripping and down-stream processing (Dreiblatt, 2003). Each of
Manufacturing Practice (cGMPs) norms. these steps can affect the properties of the final extrudates.
238 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

Feeding of extruders can be either in the “starve-fed” or the in line to ensure final desired product attributes. Using PAT as an
“flood-fed” mode. For pharmaceutical processing, feeding is com- analytical tool, flow properties, polymer structures, polymer–drug
monly conducted in a “starve-fed” mode, which results in efficient interactions as well as concentrations of drugs and additives can
mixing of the feed material as opposed to flood feeding. Starve feed- be determined immediately downstream of the extrusion process.
ing primarily uses gravimetric or volumetric feeders to dispense Several analytical techniques (rheometric, optical, ultrasonic, elec-
the material directly into screws, that prevents the accumulation trical, spectroscopy (Raman and Near-infrared (NIR) spectroscopy)
of the feed material at the feed zone and thus the mass flow rate is have been successfully incorporated into PAT for pharmaceutical
independent of screw speed. At steady state, in a starve-fed mode, manufacture. However there are limited reports of applications of
the mass flow rate at the feed zone is equal to the mass exiting the PAT in the pharmaceutical melt extrusion process (Saerens et al.,
barrel and thus accumulation in the barrel is negligible. However, 2011, 2012), indicating that the true potential of this analytical
screw speed can have a significant influence on the residence time methodology has not yet been fully realized.
distribution of the feed material (Rauwendaal, 2001).
The different zones of the barrel are pre-set to specific tempera-
tures prior to the extrusion process. The material is introduced into 3.5. Processing parameters and scale-up
the feed section of the extruder via a hopper. Ideal flow properties of
the feedstock are controlled by the angle of the feed hopper, which Scaling-up in ME like other manufacturing technologies, is an
normally exceeds the angle of repose of the feed material. Other important subject of discussion and review (Lowinger, 2011). Guns
factors such as moisture content, aeration, material density, and et al. (2012) discuss scale-up aspects of melt extrusion and make
morphology also affect the feeding properties. The feedstock is fur- a comparative assessment of miconazole and Kollicoat® IR solid
ther transported along the length of the barrel, where it is melted, dispersion prepared by laboratory scale and pilot scale extrusion
plasticized, mixed, and compressed. The shear forces created by respectively. The properties of the final dosage forms are indeed
rotating screws as well as the heating elements in the barrel gener- affected by the processing parameters (screw speed, processing
ate the required thermal energy. Mixing influences the properties temperature, and feed rate) of the melt extrusion process. The
of the final product and is either distributive mixing or dispersive screw speed and feed rate are also related to shear stress, shear
mixing. Distributive mixing refers to the content uniformity of a rate, and mean residence time, which in turn affect the disso-
particular ingredient (e.g. the API or plasticizer) while dispersive lution rate and stability of the final products. Certain minimum
mixing relates to the size reduction and distribution (Repka et al., processing temperatures are required in the ME process to reduce
2002). the torque needed to rotate the screw. Typically, the temperature
of the melting zone is set 15–60 ◦ C above the melting point of
3.3. Screw design and configuration semi-crystalline polymers or the glass transition temperature of
amorphous polymers (Mccrum and Buckley, 1997; Rauwendaal,
The efficiency of the melting process is directly related to 1994). The torque is directly proportional to the viscosity of the
the polymer’s properties and the extruder design. The screw ele- molten feedstock. The viscosity of the polymer at a fixed shear
ments can be functionally categorized into zoning, mixing and rate can be expressed by the Arrhenius equation (Repka et al.,
conveying elements respectively (Steiner, 2003; Thiele, 2003). Each 2002):
of these elements affects the characteristics of the final prod-
uct. The materials, while being mixed by the force generated  = K  × eEa /RT (2)
within a mixing element, are transported from the feeding zone
to the downstream regions by the conveying elements. There where  is the viscosity of the polymer melt, K is a constant depend-
are limited reports, however, on the effect of screw design and ing on the structure and the molecular weight of the polymer; Ea
configuration on hot melt extrusion processing. One such exam- is the activation energy of the polymer for the flow process and is
ple is the study of kneading paddle elements of the twin-screw a constant for the same type of polymer; R is the gas constant; and
extruder. Nakamichi et al. (2002) demonstrated that the kneading T is the temperature in Kelvin in Eq. (2).
paddle elements play an important role in changing the crys- As described earlier, during early stage formulation develop-
tallinity and dissolution properties of a solid dispersion of kneaded ment, the processing parameters provide the necessary design
nifedipine–hydroxypropylmethylcellulose phthalate. Verhoeven space for subsequent scale up. Additionally, stringent control
et al. (2008) described that the distribution homogeneity and the of processing parameters in turn provides a robust formulation
release rate of metoprolol tartrate in ethyl cellulose mini-matrices while maintaining the quality and increasing the production effi-
were not significantly affected by the number of kneading mix- ciency. Residual crystallinity, torque, and degradation are the
ing zones or their positions along the extruder barrel and that at common dependent variables. Meanwhile, using design of exper-
least one kneading/mixing zone was required for homogenous dis- iment (DOE), the effect of a single processing parameter on
tribution. Liu et al. studied the effects of screw configuration on formulation as well as the interplay between formulation and
the dissolution behavior of indomethacin in Eudragit EPO melt processing parameters has been studied (Verreck et al., 2003). Shi-
extrudate. Further they identified that that the kneading blocks bata et al. identified critical process parameters (residence time,
accelerated the complete dissolution process of indomethacin in screw rotation speed and heating temperature) that play an impor-
polymer melt (Liu et al., 2011). tant role in the preparation of indomethacin–crospovidone solid
dispersion by twin-screw extrusion or kneader (Shibata et al.,
3.4. Process analytical technology (PAT) 2009).
Liu et al. studied the effects of extrusion process parameters
Process analytical technology (PAT) has gained much atten- (mixer temperature, screw rotating speed and residence time)
tion and application in the pharmaceutical industry (Gnoth et al., on the dissolution behavior of melt-extruded indomethacin in
2007; Rathore et al., 2010; Read et al., 2010; Rodrigues et al., 2006; Eudragit E PO. It was observed that an increase in the mixer tem-
Scott and Wilcock, 2006; Yu et al., 2004) and has been encour- perature or the screw rotating speed, related to the corresponding
aged by FDA over the last decade (Food and Drug Administration, increase in dissolution rate. Also, the residence time for an extru-
2004). PAT can be used as a tool to monitor, analyze, and char- sion process and the drugs solubilization time in molten polymer
acterize melt extrusion and chemical composition of the products were found to be in a similar range (Liu et al., 2010).
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 239

3.6. Screening criteria

The physical and chemical properties of APIs and carriers need


to be carefully assessed prior to developing melt-extruded formula-
tions. For example, the aqueous solubility and melting point of the
drug substance and the glass transition temperature of the poly-
mers will affect the miscibility or solubility of the drug substances in
the solid dispersion systems, which will in turn affect stability. The
other properties of APIs and polymers that affect the final formu-
lation include but are not limited to hydrogen-bond acceptor and
donor groups, molecular weight, hydrophobicity, log P, and Tm /Tg
ratio. A comprehensive discussion on selection criteria for suitable
polymers could be referred to for further details (Shah et al., 2012).
Solubility parameters may be calculated to predict the mis-
cibility of the drug/polymer systems. The dispersive, polar and
hydrogen bonding moieties of drug and polymer structures con-
tribute to the results of solubility parameters (Eq. (3)). It has been
advocated that if the differential value between drug and polymer
is less than 7.0 MPa1/2 , the system is most likely miscible, while dif-
ferential value greater than 10 MPa1/2 is indicative of immiscibility
(Greenhalgh et al., 1999).

Solubility Parameter Equation : ı2T = ı2d + ı2p + ı2h (3)

The lattice based Floury–Huggins equation was originally used to


study the polymer–solvent or polymer–polymer miscibility based
on Gibbs free energy change before and after mixing. However, it
was discovered that it may also be suitable for small molecules
and polymer systems (Zhao et al., 2011). An attempt was made to
determine interaction parameter () of the model system at two
different conditions with the utilization of Nishi–Wang equation
based on melting point depression data (Nishi and Wang, 1975) and
Hildebrand and Scott correlation (Hildebrand and Scott, 1950) with
solubility parameter respectively. Temperature-composition phase Fig. 3. (a) Dissolution of melt extruded solid dispersions of paclitaxel and pure durg
diagram generated a spinodal curve, which serves as a boundary and (b) FT-IR studies of extrudates, physical mixture, excipients and paclitaxel.
between the unstable and metastable regions. Below this curve, Reprinted with Permission of The AAPS Journal (Lu et al., 2011), Copyright (2011).
the system would spontaneously form two phases.
succinate (HPMCAS), the solubility of paclitaxel was maintained
4. Solubility enhancement using hydrophilic polymers up to 12 h. FT-IR studies were conducted to determine the inter-
actions between polymer and drug, which it is believed to be the
Hydrophilic carriers, including polyvinylpyrrolidone and possible mechanism of precipitation inhibition. The stretch peak
polyvinylpyrrolidone-co-vinyl acetate, polyethylene gly- of the O H group in pure paclitaxel at 3468.02 cm−1 as well as the
cols, polyethylene oxides, some celluloses, polymethacrylate stretch peak of the carbonyl group at 1703 cm−1 were absent in
derivatives (most of which find application in enteric drug the spectrum of the extrudates, which confirmed the formation of
delivery systems), and a polyvinyl caprolactam–polyvinyl hydrogen bonding with HPMCAS (Fig. 3a) (Lu et al., 2011).
acetate–polyethylene glycol graft copolymer have been suc- Numerous studies have been conducted on the solu-
cessfully used to improve the solubility and hence bioavailability bility enhancement of water insoluble compounds using
of water insoluble compounds using melt extrusion techniques polyvinylpyrrolidone (PVP) or its derivatives as a carrier. Fu
by the solid dispersion mechanism (Table 1). The selection of et al. prepared a nimodipine–PVP VA solid dispersion tablet using
a suitable carrier mainly depends on the following factors, the hot melt extrusion technology. The dissolution profile of ME
solubility/miscibility of drug–polymer systems, the polymer tablets was compared with the commercial product Nimotop® (Fu
physico-chemical properties, stability, as well as the prerequisites et al., 2010). The melt extruded tablets exhibited a faster release
of the final dosage forms. over Nimotop® , however, the ME tablets demonstrated a reduced
McGinity and Koleng (1997) prepared acetaminophen (APAP) dissolution profile after 20–30 min, indicating a recrystallization
hot melt extruded granules with low molecular weight polyethy- behavior due to supersaturated nimodipine in the dissolution
lene glycol (PEG), which were later compressed into tablets. 80% medium. The bioavailability of ME tablets were further investi-
APAP was released within 30 min from the tablets containing gated in beagle dogs and the solid dispersion tablets illustrated a
melt extruded granules with 15% PEG. Rubessa and co-researchers similar Cmax and AUC as Nimotop® . A physically and chemically
investigated the possibility of PEG 4000 as a potential rapid release stable solid dispersion of a new chemical entity manufactured
carrier for a poorly water-soluble compound, carbamazepine. The by Novartis Pharmaceuticals Corp. was developed by Lakshaman
extrudates demonstrated a more rapid release compared to the et al., using polyvinylpyrrolidone K30 (PVP-K30) as a carrier
physical mixture (Perissutti et al., 2002). Lu et al. developed a (Lakshman et al., 2008). The water insoluble API was converted
supersaturated paclitaxel formulation using polyethylene oxide to an amorphous state and the formulation demonstrated a
(PolyOx® N80) as the primary carrier. The solubility of paclitaxel more rapid release compared with the control formulation under
was increased up to 9-fold compared to the pure drug. How- non-sink conditions. Furthermore, both of the melt extruded
ever, with the addition of hydroxypropylmethylcellulose acetate solid dispersions containing 20 and 30% drug loading dispersions
240 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

Table 1
Hydrophilic polymers for solubility enhancement.

Chemical Name Trade name Tg (◦ C) Tm (◦ C)


®
Polyvinyl Soluplus 70 –
caprolactam–polyvinyl
acetate–polyethylene
glycol graft
copolymer
Polyethylene oxide PolyyOx® WSR −57 to −50 62–67
Poly(vinyl Kollidon® , Plasdone® 90–156 –
pyrrolidone)
Polyvinylpyrrolidone- Kollidon® VA64 101 –
co-vinyl
acetate
Poly(dimethylaminoethylmethacrylate- Eudragit® E 50 –
co-methacrylic
esters)
Hydroxypropyl Aqoat-AS® ∼120 –
methylcellulose
acetate
succinate
Hydroxypropyl KlucelTM Softens at 130 ◦ C Chars at 260–275 ◦ C
cellulose
Hydroxypropyl Methocel® , Benecel® 160–210 –
methylcellulose

Polyehtylene glycol Carbowax ®
−17 C for MW 6000 37–63
Polyvinyl Kollicoat IR® 45 208
alcohol–polyethylene
glycol copolymer
Poly – –
(lactic-co-glycolic
acid) (PLGA)
Cyclodextrin CAVAMAX®
Poloxamer Lutrol® F 127 – 55

demonstrated a significantly higher bioavailability compared with temperatures and relative humidities (4 ◦ C, 25 ◦ C/60%RH, 30 ◦ C,
the control formulation containing 20% crystalline drug triturated 70%RH, 40 ◦ C/75%RH, and 50 ◦ C) (Verreck et al., 2003). A compara-
with poloxamer in this dog model. The 20% drug loading solid tive study described the use of three HPMC based polymers (HPMC
dispersion was selected for further studies and was proven as 3cps, HPMC phthalate (HPMCP) and HPMC acetyl succinate (HPM-
physically and chemically stable at controlled room temperature. CAS)) as carriers in ME based solid dispersions. Solid dispersions of a
Other types of widely used polymers in melt extrusion are water insoluble drug, NVS981 were prepared using the HPMC based
cellulose and its derivatives, including hydroxypropyl cellulose polymers at drug loadings of 20 and 50%, respectively. The HPMC
(HPC), hydroxypropyl methylcellulose (HPMC, hypromellose) and 3cps and HPMCAS demonstrated better extrudability compared to
hydroxypropylmethylcellulose acetate succinate (HPMCAS). A HPMCP (partially due to the lower Tg and viscosity), However, the
recent study from Repka’s research group illustrated the util- NVS981–HPMCP exhibited a considerably higher release rate, even
ity of low molecular weight hydroxypropyl cellulose, KlucelTM under non-sink conditions, which was attributed to the possible
ELF hydrophilic matrices for the solubilization of poorly soluble strong interactions between the polymer and the drug (Ghosh et al.,
drugs. The immediate release ME tablets of the poorly water sol- 2011).
uble drug, ketoprofen (KPR), using KlucelTM ELF as a carrier were
found to exhibit similar release profiles as commercially available
50 mg capsules of ketoprofen. Therefore, the results demonstrated
KlucelTM ELF as a potential alternative carrier when produced by
ME (Mohammed et al., 2011). Moreover, different grades of low
molecular weight HPC polymers including KlucelTM ELF and EF
were utilized to improve the dissolution rate of a low Tg model drug,
fenofibrate. The dissolution rate of fenofibrate was significantly
increased compared to the pure drug (p < 0.05). Incorporation of
sugars within the formulation was observed to further benefit the
release of fenofibrate (Fig. 4). In addition, polyvinylpyrrolidone
17PF and amino methacrylate copolymer (Eudragit® E PO) exhib-
ited a significant inhibitory effect on fenofibrate recrystallization
in the melt extrudates serving as a stabilization agent within the
formulations (Deng et al., 2012). Verreck and co-workers prepared
itraconazole–HPMC solid dispersions using hot melt extrusion
coupled with a DOE. With the addition of DOE, the processing
parameters were fully investigated, including temperature, feed
rate, and screw speed. The amorphous solid dispersion showed
Fig. 4. Dissolution of fenofibrate from the pellet formulations. Pellets were filled
a significant enhanced dissolution rate compared to the physi- into capsules so as to yield 25 mg fenofibrate per capsule.
cal mixture. The formulation was also confirmed to be physically Reprinted with Permission of Informa Healthcare USA, Inc. Drug Development and
and chemically stable during 6 months of storage at different Industrial Pharmacy (Deng et al., 2012), Copyright (2012).
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 241

Polymethacrylate derivatives have also been widely used in melt when exposed to higher temperature and shear forces, which
extrusion to improve the solubility of water insoluble compounds. theoretically affect the stability of solid dispersions, Kollicoat® IR
Gryczke et al. utilized Eudragit® EPO with suitable excipients could be alternative carriers for selected drugs in melt extrusion
and ME techniques to produce a fast disintegrating taste-masked applications.
dosage form with improved palatability and superior patient com- Pharmaceutical scientists are also investigating the utilities of
pliance (Gryczke et al., 2011). In this study, Ibuprofen (IBU—a other excipients as carriers in solid dispersions prepared by melt
bitter active) and Eudragit® EPO were processed using ME to pro- extrusion. Among those efforts, Kleinebudde and Yano success-
duce a solid dispersion and milled to produce granules. These fully extruded indomethacin (IDM) with cyclodextrin at a ratio of
granules were blended with superdisintegrants at varying ratios 1:1 below the melting point of indomethacin, and the extrudates
and directly compressed to produce orally disintegrating tablets, demonstrated a significantly higher release rate than the physical
which had physical properties similar to the marketed Nurofen® mixture (Yano and Kleinebudde, 2010). Upadhye et al. conducted
Meltlet. The taste intensity evaluation of the extruded formulations another study utilizing cyclodextrins. The researchers prepared a
in healthy human volunteers revealed an excellent taste mask- solid dispersion of 9 -tetrahydrocannabinol hemisuccinate (THC-
ing ability with zero degree of bitterness and smoother mouth HS), an ester prodrug of 9 -tetrahydrocannabinol (THC) using
feel in comparison to the moderate roughness levels experienced two types of cyclodextrin: random methylated beta-cyclodextrin
with administration of Nurofen® fast dissolving tablets. In addi- (RAMEB) and 2-hydroxypropyl beta-cyclodextrin (HPBCD). The sol-
tion, ME tablet formulations with 25% IBU demonstrated similar ubility of THC-HS was significantly higher in RAMEB compared to
release rates as Nurofen® tablets, but 40% IBU/EPO formulations HPBCD. Thus, a combination of ME and cyclodextrin complexation
showed a faster dissolution, which was attributed to a lower talc may be used to enhance solubility, as well as stability of an API
concentration in the formulation compared to the 25% IBU/EPO for- (Upadhye et al., 2010).
mulation. Overall, the ME formulations with similar disintegration Gosau et al. developed a biodegradable poly (lactic-co-glycolic
properties and crushing strengths as that of the marketed for- acid) (PLGA)–gentamicin sulphate (GS) implant, manufactured by
mulation had faster drug release rates and could mask the bitter melt extrusion technology. Polyethylene glycol 400 (PEG 400) was
taste effectively. He et al. evaluated the pharmaceutical perfor- added as a plasticizer as well as a release modifier. The implants
mance of fenofibrate solid dispersions utilizing Eudragit® E100 as containing 25% GS demonstrated a higher release rate in the ini-
a hydrophilic carrier (drug/polymer ratio 1:2 and 1:4) (He et al., tial state followed by equivalent release profiles compared to the
2010). The solid dispersions significantly increased the release marked product Septopal® (Blomet, Dramstadt, Germany) (Gosau
rate of fenofibrate compared to the physical mixture at the same and Muller, 2010). In a US patent application by Sherry et al. orally
drug/polymer ratio. It is noteworthy that the formulations exhib- disintegrating tablets (ODT) of a non-steroidal anti-inflammatory
ited a slower release in 0.1 N HCl with the increase in the percentage drug (NSAID), paracetamol were prepared by dry blending the
of Eudragit® E100, while a higher release rate in water. This phe- drugs with sugar alcohols (xylitol, mannitol, sorbitol) and subse-
nomenon was explained by the competition of two factors, pasted quently melt extruding the mixture by heating to a temperature
clumping and the solid dispersion effect (negative and positive role above the melting point of the sugars. The extrudates obtained
in the determination of dissolution rate, respectively). The authors after cooling and solidification were milled utilizing a cone mill. The
also compared the bioavailability of solid dispersions with com- milled extrudates or granules were mixed with other formulation
mercial Lipanthyl® capsules. Eudragit® E100 solid dispersions at a components and compressed into tablets. The ODTs obtained by
drug/polymer ratio of 1:4 demonstrated a 177.1% relative bioavail- complete melting of xylitol (low melting sugar alcohol) in the phys-
ability. ical mixture were reported to be more robust compared to those
Recently, a novel polyvinyl caprolactam–polyvinyl produced by conventional dry blending processes (Sherry, 2007).
acetate–polyethylene glycol graft copolymer, Soluplus® , was Recently, Poloxamer 407 (generally used as a solubilizing agent)
specifically designed and developed for melt extrusion. This was investigated as a matrix carrier for a water insoluble, high melt-
polymer has both hydrophilic and hydrophobic elements, which ing point compound, Carbamazepine (191–192 ◦ C). In this study,
therefore enhances the ability of forming a solid solution as well the formulations containing 10–50% drug load were successfully
as increasing solubilization. Therefore, Soluplus® performs very extruded in the temperature range of 50–60 ◦ C. All of the devel-
well as a solubilizing carrier for hot melt extrusion technology. In oped formulations demonstrated a significantly higher release rate
addition, Soluplus® provides a wide range for potential candidates, compared to a control formulation under non-sink conditions (Lu
from low melting to high melting drugs, including heat-sensitive et al., 2012).
APIs, due to its comparatively low Tg (70 ◦ C). Linn et al. (2012) Kindermann et al. recently fomulated polyelectrolyte com-
studied the capacity of Soluplus® to increase intestinal absorption plexes of water insoluble acidic drugs and basic polymethacrylates
of three BCS II drugs, danazol, fenofibrate and itraconazole. All (EUDRAGIT® E PO) using hot melt extrusion technology. A tailor-
of the extrudates containing the three drugs demonstrated sig- made release profile was achieved with the addition of inorganic
nificant increases in the plasma AUC compared to the crystalline salts (Kindermann et al., 2011). It was observed that the intensi-
pure drug or the physical mixture. In addition, the results of ties of stretching vibrations of the vicinal CH2 group at 2771 and
in vitro Caco-2 transport experiments confirmed the strong effect 2823 cm−1 coresponding to the non-protonated amine group of
of Soluplus® on the enhancement of absorption behaviors of the Eudragit® E PO in naproxen-extrudates were significantly lower
model drugs. These data demonstrated the much-needed addition than the physical mixture. This phenomenon was attributed to
of an effective, extrudable polymer in the pharmaceutical formu- the drug–polyelectrolyte interaction. Dissolution studies were fur-
lators’ toolbox. The utility of Kollicoat® IR, initially developed as ther conducted in non-sink conditions, and the maximum effect of
a coating polymer, was evaluated as a carrier in solid dispersions tailor-made release was achieved when sodium chloride was added
prepared by hot melt extrusion. Itraconazole–Kollicoat® IR solid at concentrations of 0.05–0.15 M.
dispersion systems demonstrated instant release properties in Polymer blends have gained considerable attention from
simulated gastric fluid without pepsin (SGFsp ) (Janssens et al., formulation scientists for the reason that they offer several physico-
2007). Interestingly, supersaturation was maintained as long chemical attributes to the dosage form as opposed to single
as 4 h for formulations containing 15, 20 and 25% itraconazole, polymer. Kalivoda and co-researchers utilized polymer blends,
which would be expected to further benefit in vivo absorption. including copovidone (COP), polyvinyl caprolactam–polyvinyl
Although the crystallinity of Kollicoat® IR was found to increase acetate–polyethylene glycol copolymer (PVCL–PVAc–PEG) and
242 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

definition, an amorphous material is a solid material without long-


range order characteristics of a crystalline material. Analytical
characterization in amorphous solid dispersion aims at accurately
determining solid-state drug–polymer miscibility, kinetics of solid-
state phase separation and nucleation – crystallization kinetics to
further assists in setting the polymer selection criteria. However,
recently there is growing interest in the development of crystalline
solid dispersion with micro/nano size range of the crystalline drug
dispersed in the amorphous or crystalline matrix for improving
the dissolution of poorly soluble drugs that is devoid of any issues
related to physical stability of drugs in amorphous solid dispersion
(Chatterji et al., 2012; Thommes et al., 2011).
Many attempts have been made to determine the degree of
crystallinity of melt extruded formulations. Undoubtedly, this is
one of the most critical testing steps in the evaluation of a ME
processed amorphous solid dispersion formulation. Selection of
an appropriate/suitable polymer, satisfactory levels of drug load-
ing and efficient and precise process control are key determinants
Fig. 5. Dissolution profiles of CBZ, CBZ–NIC cocrystal, solid dispersions prepared by for successful amorphous formulation development. Any variation
melting method and the corresponding physical mixtures (n = 3). within these parameters can result in incomplete conversion of
Repqcience + Business Media, LLC: Pharmaceutical Research (Liu et al., 2012b), Copy- a drug from the crystalline to an amorphous state. “Trace” levels
right (2012). of crystalline drug in amorphous formulations serve as seeds for
recrystallization during in vitro and in vivo dissolution or during
hypromellose 2910/5 (HPMC), to increase the solubility of a poorly storage, which can further jeopardize the stability of the formula-
water soluble compound fenofibrate (Kalivoda et al., 2012). Solid tion and hence the entire drug development program.
dispersions were prepared using hot melt extrusion with varied Determination of residual crystallinity has been an important
polymer ratios in the blends. The dissolution studies were con- analytical characterization aspect for melt extruded amorphous
ducted in a non-sink condition containing 500 mL of hydrochloric solid dispersions. Residual crystalline drug in amorphous solid
acid medium (pH 1.2) with an addition of 0.1% polysorbate 80. dispersion can serve as nuclei for recrystallization, leading to desta-
It was observed that the melt extrudates demonstrated a signifi- bilization of the dispersion upon storage and ultimately resulting
cantly faster release rate as compared to pure the fenofibrate or in a failed formulation strategy (different dissolution and bioavail-
physical mixture. The release profiles of the developed extrudates ability profile of the crystalline drug).
were further compared with marketed formulations, Lipidil and While several analytical techniques have been applied for over-
Lipidil-Ter, and was found to exhibit a superior dissolution profile all characterization of melt extruded products, for the purpose
than the marketed formulations. Interestingly, with the addition of this review the authors will only focus on techniques provid-
of PVCL–PVAc–PEG or HPMC into the formulations, the the initial ing valuable information (including quantification) on crystallinity,
release rate and the degree of supersaturation were influenced. or amorphicity of poorly soluble drugs designed as dispersions.
A BSC Class II drug with a low Tg (4 ◦ C), 3-methoxy- Moreover, an array of such orthogonal techniques as opposed to
1,5-bis(4-methoxyphenyl)-1H-1,2,4-triazole was formulated with a single technique is imperative for successful characterization of
polymer blends containing Hydroxypropylmethylcellulose (HPMC, ME dosage forms.
TC-5R) either with Copovidone (PVP-VA, Kollidon VA64 or Liu et al. (2012a) described the evaluation of miscibility of
polyvinylpyrrolidone (PVP, PVP-K30) (Sakurai et al., 2012). The Tg indomethacin and Eudragit® EPO employing thermal, rheological
of the formulations was increased with the incoporation of PVP-VA and spectroscopic analysis. Qi et al. describe successful char-
or PVP which is possibly due to the formation of hydrogen bond- acterization of melt extruded solid dispersions employing such
ing between drug and C O groups in PVP-VA or PVP, indicating orthogonal (thermal, microscopic, and spectroscopic) analytical
an improvement in the physical stability of the extruded formula- methods (Qi et al., 2008, 2010; Qi and Craig, 2010).
tions. In vivo absorption studies (Fig. 5) of these formulations were
also performed in male beagle dogs (n = 6). The formulation of drug,
HPMC and PVP at a weight ratio of 1:1:1 demonstarted an increased 5.1. Spectroscopic techniques
solubility and bioavailability as compared to the drug–HPMC solid
dispersion, resulting from a combination effect of the oral absorp- 5.1.1. X-ray diffraction methods
tion enhancement from HPMC and stability improvement from X-ray diffraction/X-ray powder diffraction (XRD/XRPD) is one
PVP. The combination of polymer blends could enhance the prop- of the most widely used identification technique for crystalline
erties of each single polymer in one formulation, which provides pharmaceuticals. It is important to note here that XRPD detects the
an alternative to formulation development. However, the miscibil- presence of molecular order (i.e. crystalline state) and not disorder
ity of polymer–polymer and drug–polymer, the phase separation (amorphous state). The amorphous state is rather implied by the
behavior of drug in the system, the long term stability and any other absence of characteristic Bragg’s peaks and hence the associated
properties need to be thoroughly investigated. long-range symmetry order (Saleki-Gerhardt et al., 1994).

5. Characterization (i) In the context of melt extrusion, XRD/XRPD finds appli-


cation in two major areas, Product characterization during
As described previously, solubilization in ME primarily occurs design/development (e.g. screening miscibility of amorphous
by amorphous solid dispersion. An amorphous system is a dis- drug and the polymer, residual amount of crystalline or amor-
ordered system with random molecular configuration, in which phous drug in dispersion etc).
individual molecules are randomly oriented with respect to (ii) Assessment of storage stability of melt extruded systems
one another and exist in various conformational states. By (e.g. factors influencing recrystallization of amorphous solid
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 243

dispersions at defined storage conditions, phase separation in In comparison to the standard method employing Bragg’s peak area
hot-melt extruded solid dispersions). calculation, PLS analysis significantly improved the accuracy of %
drug crystallinity prediction. Additionally the PLS model enabled
XRD has been used extensively to study and characterize crys- further data analysis, including identification of outliers and non-
tallinity (or amorphicity) of several APIs in melt-extruded systems linearity, as well as provided insight into factors that were most
(DiNunzio et al., 2010b; Jijun et al., 2011; Kanzer et al., 2010; important to correlate XRPD diffractograms with % crystallinity of
Schilling et al., 2010; Sun et al., 2008). XRD has proven to be a the drug (Rumondor and Taylor, 2010).
valuable tool to assess the storage stability of amorphous sys- Nollenberger et al. employed the pair distribution function (PDF)
tems (Albers et al., 2009; Andrews et al., 2010; Maclean et al., analysis coupled X-ray powder diffraction (XRPD) to determine
2011; Prodduturi et al., 2007; Thommes et al., 2011). Co-crystals of local structural order in amorphous melt extrudates of felodipine
Ibuprofen and Nicotanimide prepared utilizing hot melt extrusion with EUDRAGIT® E and EUDRAGIT® E/NE respectively. Extrudates
were evaluated by XRD. In this study it was observed that screws containing 5% EUDRAGIT® NE demonstrated a faster dissolution
with intense mixing elements at high temperatures and low mate- rate and less recrystallization in bio-relevant media. PDF analysis
rial residence times resulted in a maximum intensity XRD spectra, revealed that addition of EUDRAGIT® NE to EUDRAGIT® E during
thereby confirming formation of co-crystals (Dhumal et al., 2010). extrusion changed the local structure of EUDRAGIT® E, implying
While, XRPD is undoubtedly an indispensible qualitative identi- that the local structure is an important drug release criteria in
fication tool for crystallinity/amorphicity, quantitative application amorphous systems. Unlike the other standard analytical methods
is debatable. Using sucrose as a model solid, Saleki-Gerhardt et al. employed, PDF analysis could detect order differences between the
describe the application of XRD in quantification of disorder in different polymer extruded systems studied. PDF analysis revealed
pharmaceutical drugs. Using predetermined mixtures of crystalline that addition of EUDRAGIT® NE to the main component EUDRAGIT®
and amorphous samples, disorder, (or amorphicity) of the sample E during extrusion changed the local structure of EUDRAGIT® E
was determined. The limit of detection for this approach was about in a non-additive way (Nollenberger et al., 2009). Taken together,
10% (Saleki-Gerhardt et al., 1994). the use of XRPD coupled with computational methods (PDF) is a
Byard et al. (2005) describe the assessment of solid state promising means to provide better insights into the local structure
nuclear magnetic resonance (ssNMR), XRPD and differential scan- order in amorphous dispersions and explains the improved disso-
ning calorimetry (DSC) as methods to quantify amorphous content lution behavior of drugs from such polymer extrusion products.
in micronized pharmaceutical drug substance. Two XRPD based
approaches (measuring the crystalline peak areas and amorphous 5.1.2. IR and Raman spectroscopy
scattering respectively) were used for amorphous content estima- IR and Raman spectroscopy (vibrational spectroscopy tech-
tion. The alternative approach utilizing amorphous scattering by niques), can detect subtle differences in vibrational energy between
XRPD was found to be robust and insensitive to the variations in crystalline and amorphous states and hence employed in character-
crystalline defects and particle size of the material. This approach izing amorphous system.Verhoeven et al. utilized Raman analysis
established a direct correlation between the amorphous halo inten- as a qualitative analytical tool to determine sample homogene-
sity and the amorphous drug content in the sample which can be ity. They studied the effect of formulation and process variables
accurately used for quantification of the later in pharmaceutical on the release characteristics of metoprolol tartarate from ethyl-
solids (Byard et al., 2005). cellulose sustained-release mini-matrices produced by hot-melt
While the above-described approaches can accurately deter- extrusion. Raman anlaysis confirmed that metoprolol tartrate
mine % crystallinity (or amorphicity) for pure pharmaceutical solids was homogeneously distributed, independent of screw design
(drugs), the presence of polymer in melt extruded systems lim- and processing conditions (Verhoeven et al., 2008). Van Eerden-
its the use of XRPD. An analogous set of standards (with known brugh et al. describe the use of nanoscale-mid-infrared imaging of
crystallinity) can be prepared by mixing pure crystalline and amor- phase separation in drug–polymer blends and in binary polymer
phous standards of the drug in suitable ratios in a polymer matrix. It blends (Van Eerdenbrugh et al., 2012a,b). The molecular inter-
is important to note that such calibration standards would merely actions between celecoxib and PVP (Andrews et al., 2010) in
be physical admixtures and rather non-continuous systems. The use extruded matrices were studied using Fourier transform infrared
of such two-state calibration models to determine crystallinity in spectroscopy (FTIR) and Raman spectroscopy. Zheng et al. (2007)
an amorphous formulation is rather debatable. described the utility of FTIR spectroscopy for characterization of
Bates and co-workers have described the application of XRPD solid dispersions of Nimodipine prepared by ME. A careful evalu-
method coupled with computation of pair distribution functions ation of the FTIR spectra imply the formation of hydrogen bonds
(PDF) to characterize miscibility (between amorphous drug and between secondary amine and hydroxyl groups of drug and poly-
the polymer) in amorphous solid dispersions (Bates et al., 2006; mers (PVP/VA, EPO, HPMC), respectively (Zheng et al., 2007). Albers
Newman et al., 2008; Rumondor et al., 2009a). Newman et al. stud- et al. demonstrated the utility of FTIR to confirm the amorphous
ied three systems representing a miscible, phase separated, and nature of the poorly water-soluble drug celecoxib in solid disper-
solid nanosuspension of drug and polymer, respectively. The mis- sions of the polymethacrylate carrier Eudragit® E PO (Albers et al.,
cibility (between amorphous drug and the polymer) was assessed 2009) prepared by melt extrusion.
by subjecting the measured XRPD patterns of each system to com- Sathigari et al. described the amorphous-state characteriza-
putational analysis. Such an assessment can be made using DSC; tion of efavirenz (EFV)–polymer hot melt extruded systems. DSC,
however, it is limited by certain requirements. The individual Tg XRD, FTIR spectroscopy, and dissolution studies were employed
values (amorphous drug and polymer) generally must be about to characterize the extrudates. The amorphous nature of EFV in
10 ◦ C apart and the size of the phase separated domains must be the extrudates was confirmed by XRD and DSC studies. FTIR stud-
greater than 30 nm for DSC to predict miscibility. The size limita- ies revealed molecular interaction between the EFV and Plasdone
tion of domains (30 nm) justifies selecting solid nanosuspension as S-630, attributable to the decreased molecular mobility and stabi-
one system for this study (Newman et al., 2008). lization against crystallization (Sathigari et al., 2012).
Rumondor et al. described the use of Partial Least-Squares (PLS) Attenuated total reflectance Fourier transform infrared (ATR-
modeling in quantifying drug crystallinity in felodipine amorphous FTIR) spectroscopy is a modification of the traditional FTIR to
solid dispersions by XRPD. They found that PLS analysis signifi- overcome sensitivity issues. Utilizing ATR-FTIR, Qi et al. described
cantly improved the accuracy and prediction of % drug crystallinity. the non-homogeneous distribution of paracetamol in a Eudragit®
244 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

RSPO matrix. This type of spectroscopy proved to be very valuable Acryl-EZE® - and Eudragit® L100-55 extrudates respectively),
characterization tool for samples extruded at high drug loadings whereas crystal growth was observed after exposure to ambient
(Qi et al., 2008). storage conditions for 15–30 min. The study further explained sur-
In the present day, QbD and PAT paradigm, Raman spectroscopy face recrystallization to be possibly due to the lack of restraining
is being explored as an in-line quality control tool for the assess- pressure exerted by the polymer matrix at the tablet surface com-
ment of pharmaceutical unit operations (De Beer et al., 2008, pared to that within the tablet core (Bruce et al., 2007). Additionally
2011). Tumuluri et al. employed Raman spectroscopy as a quanti- Bruce et al. also determined the effects of varying humidity and
tative and qualitative analysis tool for characterization of hot melt talc content on the surface recrystallization of guaifenesin from
extruded filmscontaining clotrimazole and ketoprofen as model melt-extruded tablets. They observed both presence of talc and
APIs. Further, with appropriate modifications, these researchers storage at high humidity increased recrystallization of guaifenesin.
demonstrated the effective use of Raman spectroscopy to quantify However, there was no direct correlation between the amount of
API, both off-line and on-line. Raman spectroscopy being sensitive drug recrystallization and the talc concentration or rate of moisture
to changes in drug crystallinity, also finds application in charac- uptake, making surface recrystallization attributable to heteroge-
terizing amorphicity/crystallinity of the model drugs. Since Raman neous nucleation (Bruce et al., 2010b). Bruce et al. (2010a) describe
spectroscopy is sensitive to changes in crystallinity, this methodol- aqueous film coating as a mitigation strategy to reduce sur-
ogy also provides valuable information on the crytallinity of model face recrystallization. Film coated guaifenesin-containing tablets
drugs (Tumuluri et al., 2008). Similarly, Saerens et al. demonstrated showed prolonged onset time for drug recrystallization. Hypromel-
the utility of Raman spectroscopy as a PAT tool for inline quantifi- lose, a hydrophilic polymer (higher solubilization capacity for
cation of the drug as well as for the solid-state characterization of guaifenesin), was found to delay crystallization for longer periods of
drug–polymer matrix (Saerens et al., 2011). time in comparison to the more hydrophobic ethylcellulose (lower
solubilization capacity for guaifenesin).
5.2. Thermal techniques To identify and quantify amorphicity (or crystallinity), the
measurement needs to be completed before the recrystallization
5.2.1. Differential scanning calorimetry or chemical decomposition of amorphous material has started,
DSCa sensitive thermal characterization tool is commonly used which can be easily accomplished by employing a fast scan rate.
to detect phase transformations including melting, miscibility, Quantification of sample amorphicity (or crystallinity) requires
glass transitions, and re-crystallization of melt extrudates and ME measuring the important thermal transition events in the absence
based products. Since ME is a thermally intensive process, DSC is of overlapping events such as sample recrystallization or chemical
an indispensable tool to follow the thermal events and transforma- decomposition of the sample, which is achievable by employing a
tions occurring in the material during extrusion. In DSC, samples fast scan rate. High speed DSC or Hyper DSC or Fast-Scan DSC is
are heated at a constant rate and the critical temperatures at which an extension of the conventional DSC technique, which includes
the above-mentioned transitions occur are recorded. DSC subjects fast heating rates as high as 500 ◦ C/min. Conventional DSC meas-
samples to heat at a constant rate, while heat flow is continuously ures heat flow as a function of time and temperature, Hyper DSC
monitored. The temperatures at which the above-mentioned ther- allows measurement of this heat flow without overlapping ther-
mal events occur are recorded. mal events or sample degradation (Ford and Mann, 2012; Saunders
DSC is used as a primary screening technique to evaluate et al., 2004).
drug–polymer miscibility (Schilling et al., 2008), thermal transfor- Hyper DSC finds applications in characterization of polymorphs,
mations i.e. melting (Albers et al., 2009; Jijun et al., 2011; Ranzani determination of amorphicity in crystalline materials and determi-
et al., 2011; Sun et al., 2008; Thommes et al., 2010), recrystallization nation of drug solubility in polymers (formulation characterization)
stability (Jijun et al., 2011; Ranzani et al., 2011), glass transitions (Gramaglia et al., 2005; Lappalainen et al., 2006; Mathot et al.,
(Tg ) (Almeida et al., 2011; Maclean et al., 2011; Miller et al., 2008; 2002; Saunders et al., 2004). Shortened experimental times and
Prodduturi et al., 2007; Yang et al., 2011) and most importantly lower sample volumes are the typical advantages of Hyper-DSC.
to identify the amorphous nature (Andrews et al., 2010; DiNunzio Modulated DSC (MDSC) works on the same principle as conven-
et al., 2010b; Dong et al., 2008; Mididoddi and Repka, 2007; Miller tional DSC (Coleman and Craig, 1996) except for the fact that, a
et al., 2007; Sun et al., 2008) of the extruded systems (Maclean sinusoidal modulation (oscillation) is overlaid on the conventional
et al., 2011). Due to its rapid and high throughput nature, DSC has linear heating ramp to yield a heating profile in which the aver-
been used as a workhorse analytical technique to evaluate differ- age sample temperature still continuously increases with time but
ent quality attributes of melt extruded systems in tandem with not in a linear fashion. Specifically, MDSC permits separation of the
other known characterization methods (Ghebremeskel et al., 2006; total heat flow signal into its thermodynamic (heat capacity (Cp)
Janssens et al., 2007; Kanaujia et al., 2011; Qi et al., 2008, 2010; component showing melting and glass transition temperatures)
Thommes et al., 2011). DSC has also been used for rapid selection and kinetic components (e.g. crystallization, evaporation, curing).
of crystallization inhibitors (Bruce et al., 2007). Bruce et al. investi- MDSC therefore allows analysis of mixtures with overlapping ther-
gated the physical state of hot melt extruded guaifenesin tablets mal events.
containing either Acryl-EZE or Eudragit® L100-55. The authors Bruce et al. employed MDSC to understand the plasticizing effect
studied the physicochemical factors influencing crystal growth of of guaifenesin on Eudragit® L100-55 The glass transition temper-
guaifenesin in extrudates utilizing a combination of modulated DSC atures (Tg ) of melt extrudates containing guaifenesin in Eudragit®
and XRPD. L100-55 decreased in a concentration-dependent manner with Tg
This study also described the interesting application of scan- of Eudragit® L100-55 being 104.4 ◦ C and that with 20% guaifenesin
ning electron microscopy (SEM) to study the surface morphology being 51 ◦ C. This decrease in the glass transition temperature with
of extrudates and to investigate the recrystallization processes increasing guaifenesin content indicated plasticizing effects of the
(particularly the onset of recrystallization) on the surface of the drug on polymer thereby favoring the melt-extrusion processing
hot-melt extruded tablets. SEM of the intact and bisected tablets (Bruce et al., 2007). Janssens et al. employed MDSC to screen
conclusively demonstrated that crystallization was a rather sur- ternary solid dispersions containing itraconazole, PEG 6000, and
face phenomenon and higher for tablets with high amounts of HPMC 2910 E5. Their findings suggested that ternary dispersions
guaifenesin. SEM revealed absence of crystals on the newly exposed with 20% itraconazole, 15/85 of the PEG 6000, and HPMC 2910 E5
matrix surface, resulting from bisecting the tablets (for both respectively, formed completely amorphous system with superior
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 245

dissolution profile compared to binary, 80/20 of itraconazole/HPMC thermo mechanical analysis (L-TMA), nanothermal analysis (nano-
2910 E5 solid dispersion (Janssens et al., 2008). TA) and scanning thermal microscopy (SThM). Unlike conventional
thermal techniques, MTA allows localized heating with a probe
5.2.2. Isothermal microcalorimetry (IC) at high rates up to 25 ◦ C/s, thus analyzing properties (i.e. melting,
Isothermal microcalorimetry is employed for quantification of phase separation, recrystallization) in defined local areas without
amorphous content in the powdered samples (Briggner et al., 1994; thermal damage to theentire sample (Craig et al., 2002).
Buckton et al., 1995a,b). IC utilizes the simple concept of mea- Local thermal analysis (LTA) has been used for the charac-
suring heat change accompanying a physical or chemical process. terization of several multicomponent pharmaceutical systems at
Measuring such heat change and the associated energy change a micron scale (Dai et al., 2009; Harding et al., 2008; Qi et al.,
accompanying recrystallization of amorphous drugs at a particular 2010). Six et al. identified phase separation in itraconazole/Eudragit
humidity or temperature can lead to quantification of amor- E100 solid dispersions (prepared by hot-stage extrusion) employ-
phous content. While DSC along with its modifications (Hyper-DSC, ing Micro-TA. The AFM mode was used to visualize the different
MDSC) is a popular calorimetric method for characterizing amor- phases and LTA to characterize the phases. LTA could identify two
phous dispersions it has several limitations, subjecting samples separate and mixed phases of itraconazole and Eudragit® E100,
to excessively high temperatures being one of them. IC as the highlighting the potential of Micro-TA in determining phase sepa-
name suggests measures the heat energy changes in a sample as ration of the solid dispersion, these results independently verified
a function of changing humidity at constant temperature. Since the MDSC results on phase separation (Six et al., 2003).
changes in humidity can lead to recrystallization of amorphous Galop et al. describe use of micro-TA to assess drug crystallinity
phase, quantification of amorphous component is possible with in solid dispersions. LTA offered a unique advantage to probe
IC. IC is extremely sensitive technique and can detect less than 1% a selected area of the sample, thereby melting the drug locally
(w/w) amorphous content. without affecting the excipient. Micro-TA provided an insight into
Gaisford has extensively reviewed the applications of isother- understanding the poor dissolution performance of the solid dis-
mal microcalorimetry in solid pharmaceuticals (Gaisford, 2012; persion upon aging (Galop, 2005).
O’Neill and Gaisford, 2011). IC can be used to quantify amorphous Hasegawa et al. (2004) utilized a combination of electron probe
drug substances by controlling the relative humidity or relative microanalysis (EPMA) and micro-TA of to investigate the unifor-
vapor pressure in the sample ampoule. This technique, when oper- mity (homogeneous/heterogeneous dispersion) and physical state
ated accurately, can be very sensitive, detecting less than 1% (w/w) (crystalline/amorphous) of troglitazone in PVP K30 solid disper-
amorphous content. Typically, IC finds application for excipient sions.
compatibility and longer-term stability indicating assays as a com- Qi et al. characterized phase separation in aged hot-melt
plementary technique to DSC. extruded solid dispersions of felodipine and Eudragit® E PO
employing a combination of nano-thermal analysis, photothermal
5.3. Microscopic techniques FTIR microspectroscopy coupled with pulsed force mode AFM (Qi
et al., 2011). Qi et al. further classify such analytical methods as
Hot stage microscopy (HSM) and SEM are two such flagship global vs. localized characterization methods. The combined use of
visual techniques used to study the surface morphology of melt these global and local analysis has been advocated to better under-
extruded pharmaceuticals (Andrews et al., 2010; Liu et al., 2009; stand the phase separation phenomenon in HME solid dispersion
Mididoddi and Repka, 2007; Miller et al., 2007; Ozkan et al., 2009; formulations (Qi and Craig, 2010).
Reitz et al., 2008). Polarized light microscopy is another such impor-
tant microscopic technique employed in combination with several 5.5. Water vapor sorption
other analytical techniques to detect crystallinity in amorphous for-
mulation (Gupta et al., 2004; Onoue et al., 2010; Yoo et al., 2009). Water vapor sorption, though a resource intensive and tedious
Cross-polarized microscope differentiates between crystalline and technique, is employed in quantification of amorphous content
amorphous systems by measuring birefringence, moreover amor- in pharmaceutical solids (API’s). Mackin et al. described the util-
phous materials do not exhibit birefringence. ity of dynamic vapor sorption techniques in tandem with IC for
HSM coupled with polarized light microscopy showed that estimation of low levels of amorphous content. Data from both
paracetamol remained in the crystalline form on mixing with methods showed excellent agreement (±0.2% amorphous content)
molten polyethylene glycol (PEG). In addition, while cooling of and indicated that the amount of amorphous material generated
the drug–polymer matrix, a decrease in crystallinity of PEG was is extremely sensitive to small changes in the operating conditions
observed (Campbell et al., 2008). Ghebremeskel et al. (2006) (Mackin et al., 2002). Grisedale et al. (2011) studied the effects of
describe the use of polarized light microscopy in combination with milling on the structure and crystallization behavior of salbuta-
other standard analytical techniques to assess the physical stability mol sulfate using gravimetric vapor sorption analysis in addition
of melt extruded solid dispersions upon stress testing. to other standard techniques such as mDSC and XPRD.
Liu et al. used hot stage polarized optical microscopy to monitor Water vapor sorption also finds application as an independent
the co-crystal formulation. The morphological changes occurring characterization tool for solid dispersions (Konno and Taylor, 2008;
in drug–polymer blends due to heating (ambient temperature to Marsac et al., 2008). Marsac et al. describe the recrystallization of
200 ◦ C at 10 ◦ C/min) could be recorded using a camera for further nifedipine and felodipine from amorphous molecular level solid
analysis (Liu et al., 2012b). dispersions in the presence of PVP and moisture. The amount
of water sorbed, was measured using isothermal vapor sorption.
5.4. Microthermal analysis (Micro-TA) Water vapor sorption techniques have also been coupled with spec-
troscopic techniques such as Raman spectroscopy (Feth et al., 2011)
Microthermal analysis employs a combination of thermal and and Near IR spectroscopy (Moran and Buckton, 2009) for charac-
microscopy techniques providing the unique advantage of heating terization of amorphous systems.
and simultaneously imaging specific regions of the sample at sub- Dong et al. (2008) describe the use of water vapor sorption
micron resolution. technique (in addition to several other characterization tools) to
Micro-TA is a collective term encompassing a set of techniques evaluate the solid-state properties of solid dispersions prepared by
including localized differential thermal analysis (L-DTA), localized melt-extrusion and solvent co-precipitation.
246 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

Prodduturi et al. describe the use of rapid dynamic vapor co-crystal formation and its purity. Residence time decreases as
sorption technique for analysis of films containing hydrox- the screw speed increases, therefore the co-crystals produced at
ypropyl cellulose and clotrimazole. Films were investigated for slower speed demonstrated higher co-crystal purity than those
moisture-sorption, mechanical properties, and release character- produced at higher screw speeds. Even though both the batches
istics (Prodduturi et al., 2004). of co-crystals produced at 80 ◦ C and 90 ◦ C were above the eutec-
While each one of the above mentioned characterization meth- tic temperature, the co-crystal produced at 90 ◦ C showed higher
ods have individual limitations, employing a battery of such purity than that at 80 ◦ C. Of all the formulations, the co-crystals pro-
methods as opposed to a single method always provides a better duced at high shear, higher temperature (90 ◦ C) and lower screw
understanding of the system being studied. Finally, the choice of speed demonstrated pure co-crystals with no traces of un-reacted
characterization method primarily depends upon the properties components. Thus, the authors further studied dissolution and sta-
of the material (physical, thermal and chemical stability etc.) bility on the co-crystals with high purity. The dissolution studies
and the underlying phenomenon being studied (phase separation, indicated that the co-crystals showed improved release profiles
amorphicity, crystallinity etc.). Desired product quality attributes, compared to pure Ibuprofen, micronized Ibuprofen, and the phys-
confirmatory data needed, and of course, available equipment to ical mixture of Ibuprofen with Nicotinamide. In addition, these
the pharmaceutical scientist are just a few additional consider- co-crystals were found to be physically stable for up to 6 months
ations prior to design of experiments. when stored at ambient conditions.
In another study, Liu et al., investigated the in situ formation
6. Innovation in melt extrusion of co-crystals in a single-step, utilizing ME technology, in order to
achieve minimal thermal degradation of the active by extruding
6.1. Formation of co-crystals using melt extrusion the heat sensitive drugs at lower processing temperatures. In this
study, Carbamazepine (CBZ) and Nicotinamide were used as a
Every day there are numerous new chemical entities (NCEs) model drug and co-crystal former, respectively, while polyvinylpy-
being discovered, and most of these face problems of poor rolidone/vinylacetate (PVP/VA 64), Soluplus® and HPMC were used
aqueous solubility and lack of adequate physical and chemical as different carrier matrices. The active, co-former and the poly-
stability. Pharmaceutical scientists have been diligently working meric matrix were mixed together and fed through the extruder
to formulate these drugs in multiple ways by improving their to obtain a solid dispersion with in situ co-crystallization of API
physico-chemical properties, making them suitable for appropriate during the process (Liu et al., 2012b).
applications. The formation of co-crystals is considered by some The authors noticed the possibility of the drug co-crystallization
as one of the potential techniques that can achieve the desired with Nicotinamide in PVP/VA 64 matrices during DSC character-
properties of interest, such as improved dissolution rate, stability, ization of the drug–polymer physical mixtures. The hot-stage
and bioavailability (Smith et al., 2011). Co-crystals are defined as microscopy studies revealed that the co-crystals were formed
non-ionic, multi-molecular crystalline complexes. The crystal lat- completely in situ during the extrusion process, in both Soluplus®
tices of co-crystals are mainly composed of an active component and PVP/VA 64 matrices (a single melting point lower than the
and co-crystal former in a stoichiometric ratio, which are bound API); however, only partial co-crystallization occurred in HPMC
together through non-covalent interactions, primarily hydrogen matrices which was evident with melting of crystals at both, 165 ◦ C
bonding. The co-crystals formed might exhibit entirely different (melting temperature of co-crystal) and 190 ◦ C (melting tempera-
physical properties than the pure components. Many studies have ture of Carbamazepine). These results were also confirmatory with
proved that co-crystals are beneficial in improving the solubility, FTIR and XRD data, which further demonstrated that co-crystal
stability, bioavailability, and mechanical properties of the active formation, is instantaneous during melt extrusion processing in
drug. Co-crystals are mainly formed by solvent-based crystalliza- PVP/VA 64 matrices. In addition, dissolution data in PVP/VA 64
tion such as slurry, conversion, evaporation, anti-solvent addition matrix revealed that the release from co-crystals was relatively
and solid based techniques such as co-grinding and melt extru- faster than compared to the pure API. The drug release from the
sion. Of all the above-mentioned techniques, slow evaporation and physical mixtures and the drug–polymer melt extrudates was less
co-grinding are the most widely used, however both of these tech- than 80% in 2 h, and about 100% in 1 h, respectively, while the
niques possess scale up limitations. Therefore, in the recent years, solid dispersion containing API-Nicotinamide-PVP/VA64 released
significant progresses have been made to use ME technology to 100% API in just 20 min (Fig. 6), demonstrating the influence of
produce pharmaceutical scalable, solvent free and cost-effective Nicotinamide as a hydrotropic agent, and its formation of
co-crystals. co-crystals simultaneously during the extrusion process. Similar
Dhumal et al. (2010) have successfully prepared the co-crystals dissolution results were observed for the extrudates formed from
of Ibuprofen and Nicotinamide (1:1) utilizing melt extrusion tech- Soluplus® and HPMC matrices. Overall, the melting point of the
nology. In addition, these researchers have studied the effect of API was drastically reduced due to the formation of the co-crystal,
different process parameters such as screw configuration (low which helped in extruding the drug incorporated polymeric solid
shear, medium shear and high shear), screw speed (20, 30, and dispersions at lower temperatures. The processing temperatures
40 rpm) and extrusion temperature (70, 80 and 90 ◦ C) on forma- could also be adjusted based on choice of the co-former employed
tion of co-crystals and its purity. Preliminary DSC studies have during co-crystallization; however, the effect of co-former on
reported that the eutectic temperature for Ibuprofen and Nico- drug re-crystallization, physical stability, moisture sorption prop-
tinamide was 74 ◦ C, therefore extrusion temperatures above and erties, as well as other parameters, should be considered for the
below the eutectic temperature were selected to study their effect production of a stable formulation.
on co-crystallization. XRPD and DSC studies were carried out to A few examples of co-crystal complexes that were successfully
characterize the crystallinity of the final products produced from formed utilizing ME technology are Ibuprofen–Nicotinamide
ME. The characteristic XRPD peaks of Ibuprofen and co-crystals (Dhumal et al., 2010). Daurio et al. (2011) describe twin-screw
of IBU:NIC (1:1) was found to be at 2 = 6◦ and 3.1◦ , respectively. extrusion as an effective, environment-friendly, and scalable
The formulations extruded at temperatures above the eutectic method for producing co-crystals of Caffeine-Oxalic acid, Nic-
temperature (80 and 90 ◦ C) were found to have high co-crystal otinamide trans Cinnamic acid, Carbamazepine-Saccharin, and
purity. In addition, at the studied temperatures above the eutec- Theophylline-Citric acid. The authors identified this neat, and
tic temperature, screw configuration exhibited a major role in the liquid-assisted melt extrusion technique as a viable alternative
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 247

potentially degrade both the API and the carrier matrix. For thermo-
sensitive drugs, to perform the extrusion process at relatively lower
temperatures, the process may be combined with other techniques
such as rotary evaporation, spray-drying and spray-granulation,
where the API is initially converted into an amorphous form (to
lower the glass transition temperature). This amorphous API is later
extruded using a suitable polymeric carrier matrix by employing
lower processing temperatures to form a stable solid dispersion.
In a research, study performed by Lakshman et al. (2008) a crys-
talline, high melting NCE with poor water solubility was utilized
as a model drug and the authors formulated a stable solid dis-
persion using a combination of solvent evaporation with ME. The
authors produced an amorphous drug utilizing a solvent evapora-
tion technique, where the drug was dissolved in a large amount
of organic solvent and eventually evaporated and dried. The pro-
duced API form had demonstrated a glass transition temperature of
Fig. 6. Blood concentration of the drug after oral administration of solid dispersions
(1:1, w/w drug:polymer) with PVP-VA (䊉), PVP () and HPMC (). Mean and S.E. of around 125 ◦ C, which is much lower than the original API melting
six animals. Each solid dispersion contained to 100 mg of the drug. point (170 ◦ C). In order to form a stable solid dispersion, a suit-
Reprinted with Permission of The Pharmaceutical Society of Japan: Chemical and able carrier (PVP K30) was identified using hot-stage microscopy
Pharmaceutical Bulletin (Sakurai et al., 2012), Copyright (2012). that demonstrated complete miscibility of the API in the poly-
meric matrix over the temperatures used during extrusion. The
to the solution crystallization for formation of in situ co-crystals, polymeric solid dispersions formed did not demonstrate drug
where the temperature and the extent of mixing were indicated re-crystallization throughout the length of stability study, how-
as the primary parameters that influenced co-crystals formation ever, phase separation of drug-rich and polymer-rich phases were
(Daurio et al., 2011). noticed (two glass transition temperatures were observed) after
6 months, especially at accelerated conditions. This issue could
6.2. Fusion of solvent evaporation/spray drying techniques with be potentially lowered or avoided by utilizing proper packaging
melt extrusion conditions.
Addition of a plasticizer to the formulation lowered the torque
As mentioned previously, bioavailability of poorly soluble drugs during the melt extrusion process, which minimized the localized
can be enhanced by converting the active pharmaceutical ingre- heating of the drug and polymer, further improving the stability and
dients (APIs) into a high-energy amorphous state. However, a processability of the solid dispersion. Moreover, the pharmacoki-
problematic issue is retaining their high-energy states for longer netic data also revealed that the bioavailability of the dispersion
periods of time to improve dissolution rate, and/or bioavailabil- was significantly higher in comparison to the formulation with
ity, which could be due to supersaturated levels of API, and higher crystalline drug triturated with Pluronic. In spite of stable formula-
mobility at the molecular level. The most common method of pre- tions being developed in the current study, using such techniques
serving this high-energy state is to stabilize kinetically the drug to produce amorphous APIs prior to ME could pose potential manu-
substance throughout the product’s shelf life. Several factors such facturing and scale-up issues due to the requirement of unfriendly
as glass transition temperature of the amorphous product formed, environment organic solvents thereby limiting the use of such
viscosity of the melt, storage temperature of the product, plas- techniques. However, techniques such as lyophillization could be
ticization of the matrix and atmospheric moisture could greatly combined with ME as a potential alternative to overcome these
influence the stability of the amorphous dosage forms. One of the limitations.
common approaches identified to protect and physically stabilize
the amorphous solids from their re-crystallization tendency is to 7. Marketed formulations
disperse the material in the polymeric carriers using melt extrusion.
However, this process requires higher processing temperatures There are various solid dispersion systems as pharmaceutical
especially where the melting point of the API is higher, which could products on the market (Breitenbach, 2002), which include:

Table 2
Currently marketed and developed drug products produced utilizing hot melt extrusion technology (DiNunzio et al., 2012).

Product Indication HME purpose Company

Lacrisert® (Opthalmic Insert) Dry eye syndrome Shaped system Merck


ZoladexTM (Goserelin Acetate Prostate cancer Shaped system AstraZeneca
Injectable Implant)
Implanon® (Etonogestrel Implant) Contraceptive Shaped system Organon
Gris-PEG (Griseofulvin) Anti-fungal Crystalline dispersion Pedinol Pharmacal Inc.
NuvaRing® (Etonogestrel, Ethinyl Contraceptive Shaped system Merck
Estradiol depot system)
Norvir® (Ritonavir) Anti-viral (HIV) Amorphous dispersion Abbott Laboratories
Kaletra® (Ritonavir/Lopinavir) Anti-viral (HIV) Amorphous dispersion Abbott Laboratories
Eucreas® (Vildagliptin/Metformin HCl) Diabetes Melt granulation Novartis
Zithromax® (Azythromycin Anti-biotic Melt congeal Pfizer
enteric-coated multiparticulates)
Orzurdex® (Dexamethasone Macular edema Shaped system Allergan
Implantable Device)
FenoglideTM (Fenofibrate) Dyslipidemia MeltDose® (solid dispersion) Life Cycle Pharma
Anacetrapib (Under Development) Atherosclerosis Amorphous dispersion Merck
Posaconazole (Under Development) Antifungal Amorphous dispersion Merck
248 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

Griseofulvin–Polyethyleneglycol dispersion (Gris-PEG® marketed

Supercritical anti-solvent (SAS) (Lim et al., 2010; Uzun et al., 2011) overcomes process
by Wander) and Cesamet® , a Nabilone-PVP preparation (marketed
by Lilly). Another formulation of Troglitazone (Rezulin® ) by Parke-

limitations and produces dried composites suitable for subsequent processing.


Davis was earlier on the market, but was withdrawn from the

Residual solvent content, safety concerns; secondary drying process required


US market in 2000 due to toxicology related issues of the active.

Residual solvent in the final product (water content); stability concerns


Apart from these, several commercialized implants containing

Employs volatile organic solvents (process and environmental hazard)


Luteininzing hormone releasing hormone (LHRH) agonists such

Lack of process control, phase separation due to poor cooling rates,

Limited drug/polymer solubilities in supercritical carbon dioxide,


as Goserelin (Zoladex® ), Buserelin (Depot–Profact® ), and devices
such as Implanon® and Nuvaring® are also available in the form

High thermal stress; not suitable for thermolabile materials,


of solid dispersions/solutions. Although there is a huge potential

Material should have optimal thermoplastic properties


of formulating many poorly soluble drugs into solid dispersions

Drug and polymer require a common volatile solvent


Drug and matrix should be compatible and mix well

Phase separation upon storage; potential instability


using melt extrusion for improved bioavailability, to date, few
have been commercialized so far. This, however, is changing and
will continue to do so.

Heat intensive process; thermal instability

Energy intensive manufacturing process


A classic example for a marketed ME pharmaceutical product

High cost, time and energy inefficient,


Congealing due to insufficient drying,
is Meltrex Kaletra® tablets, which was developed by SOLIQSTM

Problem with clogged nozzle heads


(Germany) to improve patient compliance and hence their adher-
ence to the highly active anti-retroviral therapy (Breitenbach,
2006). Kaletra® is a protease-inhibitor combination product, an

High manufacturing cost


important treatment option for patients suffering from HIV, and
each of these tablets consists of 200 mg of Lopinavir and 50 mg
Ritonavir in PVP/VA matrix, as opposed to Kaletra® soft gel
capsules containing 133 mg and 33 mg of Lopinavir and Ritona-
vir, respectively. The application of melt extrusion technology
has turned scientific challenges into patient benefits, reformulat-

Cons
ing a product with many desired attributes: (a) better stability
at room temperature, (b) administration independent of meal
times, and (c) requiring fewer numbers of tablets to be admin-
istered (4 tablets/day) in comparison to 6 soft gel capsules/day
(Breitenbach, 2006). Due to these added benefits, Kaletra® was
granted a fast track approval by U. S. Food and Drug Adminis-

Lower processing temperatures compared to HME and fusion method


Rapid, high purity and high yield, carbon dioxide is safe, inexpensive
tration (FDA) in October 2005, followed by EU approval in July

and commercially available, used for drugs susceptible to hydrolytic

Continuous, rapid, robust, scalable process, can be used to produce


2006. SOLIQSTM has also demonstrated an added advantage of

Continuous, customizable, modular, robust and scalable process


Pros and cons of melt extrusion in comparison to other processing techniques for preparation of solid dispersion.

MeltrexTM technology in producing stable amorphous final dosage

Large scale production equipment available and standardized


Robust and mild production conditions (heat sensitive drugs)
forms, based on years of studies, thus eliminating issues associated
with polymorphism of drug substances, such as stability and/or
degradation-potential advantage over freeze drying

bioavailability.

Amenable to large scale commercial manufacture


In addition to the above product, SOLIQSTM has also devel-
oped a sustained release formulation of Verapamil in polyethylene

Large scale commercial production feasible


glycol–glyceride matrix (Isoptin SRE-240) that was the first directly
Simple laboratory processing equipment

Controlled high temperature processing

shaped ME product on the market, as well as a fast-onset Ibuprofen


Traditional, simple very first method

system (Andrews et al., 2010). Also, in 2010 Abbott announced that


the U. S. FDA approved a new thermally stable tablet formulation
micro and nano particulates
of the protease inhibitor Norvir® (Ritonavir). This new formulation
does not require refrigeration and can be stored at room tempera-
Solvent free processing

ture making it more convenient for patients.


Solvent free method

Few formulations that are currently under development from


Pharmaceutical companies such as Merck and Particle sciences
include Anacetrapib, Posaconazole (antifungal), Dapivirine (anti-
viral) and Maraviroc (anti-viral) etc. Anacetrapib, a cholesteryl ester
Pros

transfer protein (CETP) inhibitor, is currently being developed for


atherosclerosis by Merck as a melt extruded formulation contain-
ing Kollidon® VA 64 as a polymer matrix with vitamin E TPGS
and additional surfactant materials (DiNunzio et al., 2012). The
data from the pre-clinical studies in Rhesus monkeys demonstrated
Supercritical fluid processing (SFP)

superior bioavailability and reduced variability of the Anacetrapib


melt extruded formulation in comparison to its spray-dried disper-
sion. This ME formulation is currently being evaluated in Phase-III
clinical trials, and the early results from Phase IIb (efficacy and tol-
erability) that was presented at the American Heart Association
meeting in 2010 demonstrated that a 100 mg dosage decreased
Fusion method

Melt extrusion
Freeze drying

LDL by 36% and increased HDL by 138%, with no association with


Spray drying
Technique

increased cardiovascular disease events (Cannon et al., 2010). Addi-


tionally, Posaconazole is another solid dispersion product, which is
Table 3

being developed by Merck. The active is dispersed in a matrix of


hydroxypropyl methyl cellulose acetate succinate (HPMCAS), and
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 249

the eutectic mixture formed allows reduced temperatures for pro- Acknowledgments
duction of the product using a twin-screw co-rotating extruder,
solubilizing the active in the polymer matrix (DiNunzio et al., 2012). This study was supported by HRSA/OFAM/DGMO grant
The obtained extrudates are then milled, mixed with extra-granular #D1DHP20294. The authors would also like to thank the Pii Center
materials, and compressed into tablets containing a 100 mg dose. A for Pharmaceutical Technology for its support of this project.
comparative pharmacokinetic crossover study in healthy human
volunteers with marketed Noxafil® demonstrated a significant References
improvement in oral bioavailability with reduced variation in fast
and fed states (minimal food effects) as well as reduced inter- Abu-Diak, O.A., Jones, D.S., Andrews, G.P., 2012. Understanding the perfor-
mance of melt-extruded poly(ethylene oxide)-bicalutamide solid dispersions:
patient variability, allowing for improved product attributes of the
characterisation of microstructural properties using thermal, spectroscopic and
formulation. drug release methods. J. Pharm. Sci. 101, 200–213.
There are already several pharmaceutical products on the mar- Albers, J., Alles, R., Matthee, K., Knop, K., Nahrup, J.S., Kleinebudde, P., 2009. Mech-
anism of drug release from polymethacrylate-based extrudates and milled
ket either currently being manufactured or under development
strands prepared by hot-melt extrusion. Eur. J. Pharm. Biopharm. 71, 387–394.
using melt extrusion technology (Table 2), which demonstrates Almeida, A., Possemiers, S., Boone, M.N., De Beer, T., Quinten, T., Van Hoorebeke, L.,
its production feasibility and scaling ability for commercialization. Remon, J.P., Vervaet, C., 2011. Ethylene vinyl acetate as matrix for oral sustained
Many multi-national companies have been investing and perform- release dosage forms produced via hot-melt extrusion. Eur. J. Pharm. Biopharm.
77, 297–305.
ing extensive research with melt extrusion and are adopting this Andrews, G.P., Abu-Diak, O., Kusmanto, F., Hornsby, P., Hui, Z., Jones, D.S., 2010.
relatively new technique of producing and commercializing solid Physicochemical characterization and drug-release properties of celecoxib hot-
dispersions in recent years. melt extruded glass solutions. J. Pharm. Pharmacol. 62, 1580–1590.
Bates, S., Zografi, G., Engers, D., Morris, K., Crowley, K., Newman, A., 2006. Analysis
of amorphous and nanocrystalline solids from their X-ray diffraction patterns.
Pharm. Res. 23, 2333–2349.
8. Comparison of different solid dispersion techniques Bhattacharya, S., Suryanarayanan, R., 2009. Local mobility in amorphous
pharmaceuticals—characterization and implications on stability. J. Pharm. Sci.
98, 2935–2953.
Table 3 summarizes the pros and cons of Melt extrusion tech- Breitenbach, J., 2002. Melt extrusion: from process to drug delivery technology. Eur.
nology in comparison to other processing techniques used for J. Pharm. Biopharm. 54, 107–117.
Breitenbach, J.r., 2006. Melt extrusion can bring new benefits to HIV therapy: the
preparation of solid dispersions. It should be emphasized that no example of Kaletra® tablets. Am. J. Drug Deliv. 4, 61–64.
single processing technique or technology may provide the nec- Briggner, L.-E., Buckton, G., Bystrom, K., Darcy, P., 1994. The use of isothermal
essary means to desirable product quality attributes and hence microcalorimetry in the study of changes in crystallinity induced during the
processing of powders. Int. J. Pharm. 105, 125–135.
the successful development and commercialization of a particular
Bruce, C., Fegely, K.A., Rajabi-Siahboomi, A.R., McGinity, J.W., 2007. Crystal growth
pharmaceutical dosage form or device. formation in melt extrudates. Int. J. Pharm. 341, 162–172.
Bruce, C.D., Fegely, K.A., Rajabi-Siahboomi, A.R., McGinity, J.W., 2010a. Aqueous film
coating to reduce recrystallization of guaifenesin from hot-melt extruded acrylic
matrices. Drug Dev. Ind. Pharm. 36, 218–226.
9. Conclusion Bruce, C.D., Fegely, K.A., Rajabi-Siahboomi, A.R., McGinity, J.W., 2010b. The influ-
ence of heterogeneous nucleation on the surface crystallization of guaifenesin
Over the last two decades, the pharmaceutical industry has from melt extrudates containing Eudragit L10055 or Acryl-EZE. Eur. J. Pharm.
Biopharm. 75, 71–78.
made significant progress in the drug discovery and design front Buckton, G., Darcy, P., Greenleaf, D., Holbrook, P., 1995a. The use of isothermal
that has yielded a vast number of potential drug candidates microcalorimetry in the study of changes in crystallinity of spray-dried salbu-
with poor or limited solubility and hence limited bioavailability. tamol sulphate. Int. J. Pharm. 116, 113–118.
Buckton, G., Darcy, P., Mackellar, A.J., 1995b. The use of isothermal microcalorimetry
Melt extrusion as a manufacturing technique has been success- in the study of small degrees of amorphous content of powders. Int. J. Pharm.
fully adapted in the mainstream pharmaceutical industry and has 117, 253–256.
emerged as an innovative alternative in the drug-product develop- Byard, S.J., Jackson, S.L., Smail, A., Bauer, M., Apperley, D.C., 2005. Studies on the
crystallinity of a pharmaceutical development drug substance. J. Pharm. Sci. 94,
ment arena particularly for solubility enhancement. 1321–1335.
Melt extrusion will continue to be a highly sought alternative Campbell, K., Craig, D.Q., McNally, T., 2008. Poly(ethylene glycol) layered silicate
since there is a continuous need for innovative drug products, nanocomposites for retarded drug release prepared by hot-melt extrusion. Int.
J. Pharm. 363, 126–131.
intellectual property protection and integrated, yet cost-effective
Cannon, C.P., Shah, S., Dansky, H.M., Davidson, M., Brinton, E.A., Gotto, A.M.,
manufacturing platforms. While melt extrusion might mistakenly Stepanavage, M., Liu, S.X., Gibbons, P., Ashraf, T.B., Zafarino, J., Mitchel, Y., Barter,
be perceived to be a recent development in the pharmaceutical P., 2010. Safety of anacetrapib in patients with or at high risk for coronary heart
disease. N. Engl. J. Med. 363, 2406–2415.
industry, the first marketed melt extruded drug product using
Chatterji, A., D.D., Miller, D.A., Sandhu, H., Shah, N., 2012. Process for controlled crys-
these techniques was on the market in 1981 with several prod- tallization of an active pharmaceutical ingredient from supercooled liquid state
ucts developed thereafter or under development over the last 30 by hot melt extrusion. In: Office, U.S.P.a.T. (Ed.). United States, www.uspto.gov,
years. It is noteworthy that several products developed by melt WO/2012/110469.
Chiou, W.L., Riegelman, S., 1971. Pharmaceutical applications of solid dispersion
extrusion have successfully replaced conventional drug delivery systems. J. Pharm. Sci. 60, 1281–1302.
systems. Chokshi, R.J., Sandhu, H.K., Iyer, R.M., Shah, N.H., Malick, A.W., Zia, H., 2005. Charac-
In conclusion, producing solid dispersions by melt extrusion terization of physico-mechanical properties of indomethacin and polymers to
assess their suitability for hot-melt extrusion processs as a means to manufac-
techniques has become a popular means for improving solubil- ture solid dispersion/solution. J. Pharm. Sci. 94, 2463–2474.
ity and hence bioavailability of poorly soluble APIs. A successfully Coleman, N.J., Craig, D.Q.M., 1996. Modulated temperature differential scanning
developed melt extrusion based product is defined by several calorimetry. A novel approach to pharmaceutical thermal analysis. Int. J. Pharm.
135, 13–29.
formulation, equipment, design, and processing factors. Care- Craig, D.Q., Kett, V.L., Andrews, C.S., Royall, P.G., 2002. Pharmaceutical applications
ful assessment and control of each of these factors and their of micro-thermal analysis. J. Pharm. Sci. 91, 1201–1213.
interplay governs key product quality attributes. Finally, as poten- Crowley, M.M., Zhang, F., Repka, M.A., Thumma, S., Upadhye, S.B., Battu, S.K., McGin-
ity, J.W., Martin, C., 2007. Pharmaceutical applications of hot-melt extrusion:
tial new drug candidates (poorly soluble) continue to be discovered,
part I. Drug Dev. Ind. Pharm. 33, 909–926.
there will be an existing requirement of advanced formulation Dai, X., Reading, M., Craig, D.Q., 2009. Mapping amorphous material on a partially
development technologies. Melt extrusion will continue to be at crystalline surface: nanothermal analysis for simultaneous characterisation and
imaging of lactose compacts. J. Pharm. Sci. 98, 1499–1510.
the forefront of such formulation technologies and enable the
Daurio, D., Medina, C., Saw, R., Nagapudi, K., Alvarez-Núñez, F., 2011. Application of
commercialization of drug products, as well as unique platform twin screw extrusion in the manufacture of cocrystals, part I: four case studies.
technologies. Pharmaceutics 3, 582–600.
250 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

De Beer, T., Burggraeve, A., Fonteyne, M., Saerens, L., Remon, J.P., Vervaet, C., 2011. granules prepared by hot melt extrusion. Colloids Surf. B: Biointerfaces 86,
Near infrared and Raman spectroscopy for the in-process monitoring of phar- 275–284.
maceutical production processes. Int. J. Pharm. 417, 32–47. Grzybowska, K., Paluch, M., Grzybowski, A., Wojnarowska, Z., Hawelek, L.,
De Beer, T.R., Bodson, C., Dejaegher, B., Walczak, B., Vercruysse, P., Burggraeve, A., Kolodziejczyk, K., Ngai, K.L., 2010. Molecular dynamics and physical stabil-
Lemos, A., Delattre, L., Heyden, Y.V., Remon, J.P., Vervaet, C., Baeyens, W.R., 2008. ity of amorphous anti-inflammatory drug: celecoxib. J. Phys. Chem. B 114,
Raman spectroscopy as a process analytical technology (PAT) tool for the in-line 12792–12801.
monitoring and understanding of a powder blending process. J. Pharm. Biomed. Guns, S., Mathot, V., Martens, J.A., Van den Mooter, G., 2012. Upscaling of the hot-melt
Anal. 48, 772–779. extrusion process: comparison between laboratory scale and pilot scale pro-
Deng, W., Singh, A., Shah, S., Mohammed, N., Jo, S., Majumdar, S., Pinto, E., Tewari, D., duction of solid dispersions with miconazole and Kollicoat(R) IR. Eur. J. Pharm.
Durig, T., Repka, M.A., 2012. Stabilization of fenofibrate in low molecular weight Biopharm. 81, 674–682.
hydroxypropylcellulose matrices produced by hot melt extrusion. Drug Dev. Ind. Gupta, P., Kakumanu, V.K., Bansal, A.K., 2004. Stability and solubility of celecoxib-PVP
Pharm., http://dx.doi.org/10.3109/03639045.2012.679280. amorphous dispersions: a molecular perspective. Pharm. Res. 21, 1762–1769.
Dhumal, R.S., Kelly, A.L., York, P., Coates, P.D., Paradkar, A., 2010. Cocrystaliza- Hancock, B.C., Shamblin, S.L., Zografi, G., 1995. Molecular mobility of amorphous
tion and simultaneous agglomeration using hot melt extrusion. Pharm. Res. 27, pharmaceutical solids below their glass transition temperatures. Pharm. Res.
2725–2733. 12, 799–806.
Dierickx, L., Saerens, L., Almeida, A., De Beer, T., Remon, J.P., Vervaet, C., 2012. Harding, L., Qi, S., Hill, G., Reading, M., Craig, D.Q., 2008. The development
Co-extrusion as manufacturing technique for fixed-dose combination mini- of microthermal analysis and photothermal microspectroscopy as novel
matrices. Eur. J. Pharm. Biopharm. 81, 683–689. approaches to drug-excipient compatibility studies. Int. J. Pharm. 354, 149–157.
DiNunzio, J.C., Martin, Z.F.C., Mcginity, J.W., 2012. Melt extrusion. In: Williams III, Hasegawa, S., Hamaura, T., Furuyama, N., Horikawa, S., Kusai, A., Yonemochi, E., Ter-
R.O., Watts, A.B., Miller, D.A. (Eds.), Formulating Poorly Water Soluble Drugs. ada, K., 2004. Uniformity and physical states of troglitazone in solid dispersions
Springer, New York, NY, pp. 311–362. determined by electron probe microanalysis and microthermal analysis. Int. J.
Dinunzio, J.C., Brough, C., Hughey, J.R., Miller, D.A., Williams 3rd, R.O., McGinity, Pharm. 280, 39–46.
J.W., 2010a. Fusion production of solid dispersions containing a heat-sensitive He, H., Yang, R., Tang, X., 2010. In vitro and in vivo evaluation of fenofibrate solid
active ingredient by hot melt extrusion and Kinetisol dispersing. Eur. J. Pharm. dispersion prepared by hot-melt extrusion. Drug Dev. Ind. Pharm. 36, 681–687.
Biopharm. 74, 340–351. Hildebrand, J., Scott, R., 1950. Solubility of Non-Electrolytes, 3rd ed. Reinhold, New
DiNunzio, J.C., Brough, C., Miller, D.A., Williams 3rd, R.O., McGinity, J.W., 2010b. York.
Fusion processing of itraconazole solid dispersions by kinetisol dispers- Hughey, J.R., DiNunzio, J.C., Bennett, R.C., Brough, C., Miller, D.A., Ma, H., Williams 3rd,
ing: a comparative study to hot melt extrusion. J. Pharm. Sci. 99, 1239– R.O., McGinity, J.W., 2010. Dissolution enhancement of a drug exhibiting thermal
1253. and acidic decomposition characteristics by fusion processing: a comparative
Dong, Z., Chatterji, A., Sandhu, H., Choi, D.S., Chokshi, H., Shah, N., 2008. Evaluation study of hot melt extrusion and KinetiSol dispersing. AAPS PharmSciTech 11,
of solid state properties of solid dispersions prepared by hot-melt extrusion and 760–774.
solvent co-precipitation. Int. J. Pharm. 355, 141–149. Janssens, S., de Armas, H.N., Remon, J.P., Van den Mooter, G., 2007. The use of a
Dreiblatt, A., 2003. Process design. In: Ghebre-Sellassie, I., Martin, C. (Eds.), Pharma- new hydrophilic polymer, Kollicoat IR, in the formulation of solid dispersions of
ceutical Extrusion Technology. Marcel Dekker, Inc., New York, pp. 153–169. Itraconazole. Eur. J. Pharm. Sci. 30, 288–294.
Feng, J., Xu, L., Gao, R., Luo, Y., Tang, X., 2011. Evaluation of polymer carriers with Janssens, S., de Armas, H.N., Roberts, C.J., Van den Mooter, G., 2008. Characterization
regard to the bioavailability enhancement of bifendate solid dispersions pre- of ternary solid dispersions of itraconazole, PEG 6000, and HPMC 2910 E5. J.
pared by hot-melt extrusion. Drug Dev. Ind. Pharm. 38 (6), 735–743. Pharm. Sci. 97, 2110–2120.
Feth, M.P., Jurascheck, J., Spitzenberg, M., Dillenz, J., Bertele, G., Stark, H., 2011. Janssens, S., Van den Mooter, G., 2009. Review: physical chemistry of solid disper-
New technology for the investigation of water vapor sorption-induced crys- sions. J. Pharm. Pharmacol. 61, 1571–1586.
tallographic form transformations of chemical compounds: a water vapor Jijun, F., Lishuang, X., Xiaoli, W., Shu, Z., Xiaoguang, T., Xingna, Z., Haibing, H., Xing,
sorption gravimetry-dispersive Raman spectroscopy coupling. J. Pharm. Sci. 100, T., 2011. Nimodipine (NM) tablets with high dissolution containing NM solid
1080–1092. dispersions prepared by hot-melt extrusion. Drug Dev. Ind. Pharm. 0, 1–11.
Food and Drug Administration Process Analytical Technology Initiative, G.f.I., 2004. Kakumanu, V.K., Bansal, A.K., 2002. Enthalpy relaxation studies of celecoxib amor-
In: Administration, U.S.D.o.H.a.H.S.F.a.D. (Ed.), Process Analytical Technology phous mixtures. Pharm. Res. 19, 1873–1878.
Initiative, Guidance for Industry, PAT—A Framework for Innovative Pharmaceu- Kalivoda, A., Fischbach, M., Kleinebudde, P., 2012. Application of mixtures of
tical Development, Manufacturing and Quality Assurance. polymeric carriers for dissolution enhancement of fenofibrate using hot-melt
Food and Drug Administration Quality systems Approach to Pharmaceutical cGMP extrusion. Int. J. Pharm. 429, 58–68.
Regulations, G.f.I., 2006. In: Administration, U.S.D.o.H.a.H.S.F.a.D. (Ed.)., Quality Kanaujia, P., Lau, G., Ng, W.K., Widjaja, E., Schreyer, M., Hanefeld, A., Fischbach, M.,
Systems Approach to Pharmaceutical cGMP Regulations, Guidance for Industry. Saal, C., Maio, M., Tan, R.B., 2011. Investigating the effect of moisture protection
Ford, J.L., Mann, T.E., 2012. Fast-Scan DSC and its role in pharmaceutical physical on solid-state stability and dissolution of fenofibrate and ketoconazole solid
form characterisation and selection. Adv. Drug Deliv. Rev. 64, 422–430. dispersions using PXRD, HSDSC and Raman microscopy. Drug Dev. Ind. Pharm.
Forster, A., Hempenstall, J., Tucker Rades, T., 2001a. The potential of small-scale 37 (9), 1026–1035.
fusion experiments and the Gordon–Taylor equation to predict the suitability Kanzer, J., Tho, I., Flaten, G.E., Magerlein, M., Holig, P., Fricker, G., Brandl, M., 2010. In-
of drug/polymer blends for melt extrusion. Drug Dev. Ind. Pharm. 27, 549–560. vitro permeability screening of melt extrudate formulations containing poorly
Forster, A., Hempenstall, J., Tucker, I., Rades, T., 2001b. Selection of excipients for water-soluble drug compounds using the phospholipid vesicle-based barrier. J.
melt extrusion with two poorly water-soluble drugs by solubility parameter Pharm. Pharmacol. 62, 1591–1598.
calculation and thermal analysis. Int. J. Pharm. 226, 147–161. Khougaz, K., Clas, S.D., 2000. Crystallization inhibition in solid dispersions of MK-
Fu, J., Zhang, L., Guan, T., Tang, X., He, H., 2010. Stable nimodipine tablets with 0591 and poly(vinylpyrrolidone) polymers. J. Pharm. Sci. 89, 1325–1334.
high bioavailability containing NM-SD prepared by hot-melt extrusion. Powder Kindermann, C., Matthee, K., Strohmeyer, J., Sievert, F., Breitkreutz, J., 2011. Tailor-
Technol. 204, 214–221. made release triggering from hot-melt extruded complexes of basic polyelec-
Gaisford, S., 2012. Isothermal microcalorimetry for quantifying amorphous content trolyte and poorly water-soluble drugs. Eur. J. Pharm. Biopharm. 79, 372–381.
in processed pharmaceuticals. Adv. Drug Deliv. Rev. 64, 431–439. Konno, H., Taylor, L.S., 2006. Influence of different polymers on the crystalliza-
Galop, M., 2005. Study of pharmaceutical solid dispersions by microthermal analysis. tion tendency of molecularly dispersed amorphous felodipine. J. Pharm. Sci. 95,
Pharm. Res. 22, 293–302. 2692–2705.
Ghebremeskel, A.N., Vemavarapu, C., Lodaya, M., 2006. Use of surfactants as plas- Konno, H., Taylor, L.S., 2008. Ability of different polymers to inhibit the crystallization
ticizers in preparing solid dispersions of poorly soluble API: stability testing of of amorphous felodipine in the presence of moisture. Pharm. Res. 25, 969–978.
selected solid dispersions. Pharm. Res. 23, 1928–1936. Lakshman, J.P., Cao, Y., Kowalski, J., Serajuddin, A.T., 2008. Application of melt extru-
Ghosh, I., Snyder, J., Vippagunta, R., Alvine, M., Vakil, R., Tong, W.Q., Vippagunta, S., sion in the development of a physically and chemically stable high-energy
2011. Comparison of HPMC based polymers performance as carriers for manu- amorphous solid dispersion of a poorly water-soluble drug. Mol. Pharm. 5,
facture of solid dispersions using the melt extruder. Int. J. Pharm. 419, 12–19. 994–1002.
Gnoth, S., Jenzsch, M., Simutis, R., Lubbert, A., 2007. Process analytical technol- Lappalainen, M., Pitkanen, I., Harjunen, P., 2006. Quantification of low levels of
ogy (PAT): batch-to-batch reproducibility of fermentation processes by robust amorphous content in sucrose by hyperDSC. Int. J. Pharm. 307, 150–155.
process operational design and control. J. Biotechnol. 132, 180–186. Leuner, C., Dressman, J., 2000. Improving drug solubility for oral delivery using solid
Gosau, M., Muller, B.W., 2010. Release of gentamicin sulphate from biodegradable dispersions. Eur. J. Pharm. Biopharm. 50, 47–60.
PLGA-implants produced by hot melt extrusion. Pharmazie 65, 487–492. Lim, R.T.Y., Ng, W.K., Tan, R.B.H., 2010. Amorphization of pharmaceutical compound
Gramaglia, D., Conway, B.R., Kett, V.L., Malcolm, R.K., Batchelor, H.K., 2005. High by co-precipitation using supercritical anti-solvent (SAS) process (Part I). The
speed DSC (hyper-DSC) as a tool to measure the solubility of a drug within a Journal of Supercritical Fluids 53, 179–184.
solid or semi-solid matrix. Int. J. Pharm. 301, 1–5. Linn, M., Collnot, E.M., Djuric, D., Hempel, K., Fabian, E., Kolter, K., Lehr, C.M., 2012.
Greenhalgh, D.J., Williams, A.C., Timmins, P., York, P., 1999. Solubility parameters as Soluplus(R) as an effective absorption enhancer of poorly soluble drugs in vitro
predictors of miscibility in solid dispersions. J. Pharm. Sci. 88, 1182–1190. and in vivo. Eur. J. Pharm. Sci. 45, 336–343.
Grisedale, L.C., Jamieson, M.J., Belton, P.S., Barker, S.A., Craig, D.Q., 2011. Charac- Liu, H., Wang, P., Zhang, X., Shen, F., Gogos, C.G., 2010. Effects of extrusion process
terization and quantification of amorphous material in milled and spray-dried parameters on the dissolution behavior of indomethacin in Eudragit E PO solid
salbutamol sulfate: a comparison of thermal, spectroscopic, and water vapor dispersions. Int. J. Pharm. 383, 161–169.
sorption approaches. J. Pharm. Sci. 100, 3114–3129. Liu, H., Zhang, X., Suwardie, H., Wang, P., Gogos, C.G., 2012a. Miscibility studies of
Gryczke, A., Schminke, S., Maniruzzaman, M., Beck, J., Douroumis, D., 2011. Develop- indomethacin and Eudragit(R) E PO by thermal, rheological, and spectroscopic
ment and evaluation of orally disintegrating tablets (ODTs) containing Ibuprofen analysis. J. Pharm. Sci. 101, 2204–2212.
S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252 251

Liu, X., Lu, M., Guo, Z., Huang, L., Feng, X., Wu, C., 2012b. Improving the chemical Ozkan, S., Kalyon, D.M., Yu, X., McKelvey, C.A., Lowinger, M., 2009. Multifunc-
stability of amorphous solid dispersion with cocrystal technique by hot melt tional protein-encapsulated polycaprolactone scaffolds: fabrication and in vitro
extrusion. Pharm. Res. 29, 806–817. assessment for tissue engineering. Biomaterials 30, 4336–4347.
Liu, H., Zhu, L., Wang, P., Zhang, X., Gogos, C.G., 2011. Effects of screw configuration Perissutti, B., Newton, J.M., Podczeck, F., Rubessa, F., 2002. Preparation of extruded
on indomethacin dissolution behavior in Eudragit E PO. Adv. Polym. Technol. 31 carbamazepine and PEG 4000 as a potential rapid release dosage form. Eur. J.
(4), 331–342. Pharm. Biopharm. 53, 125–132.
Liu, L., Jin, T.Z., Coffin, D.R., Hicks, K.B., 2009. Preparation of antimicrobial mem- Prodduturi, S., Manek, R.V., Kolling, W.M., Stodghill, S.P., Repka, M.A., 2004. Water
branes: coextrusion of poly(lactic acid) and Nisaplin in the presence of vapor sorption of hot-melt extruded hydroxypropyl cellulose films: effect on
Plasticizers. J. Agric. Food Chem. 57, 8392–8398. physico-mechanical properties, release characteristics, and stability. J. Pharm.
Lowinger, M., 2011. Process development: scaling a melt extrusion process from Sci. 93, 3047–3056.
conception to commercialization. Am. Pharm. Rev., March 01. Prodduturi, S., Urman, K.L., Otaigbe, J.U., Repka, M.A., 2007. Stabilization of hot-melt
Lu, J., Shah, S., Jo, S., Repka, M.A.,2011. Solubility enhancement and precipita- extrusion formulations containing solid solutions using polymer blends. AAPS
tion inhibition of paclitaxel using hot melt extrusion technology. In: American PharmSciTech 8 (2), E152–E161 (article 50).
Association of Pharmaceutical Scientists Annual Meeting and Exposition. Wash- Qi, S., Belton, P., Nollenberger, K., Clayden, N., Reading, M., Craig, D.Q., 2010. Char-
ington Convention Center, Washington, DC, USA (abstract ID: W5246). acterisation and prediction of phase separation in hot-melt extruded solid
Lu, J., Shah, S., Repka, M.A.,2012. Solid dispersions of carbamazepine with Lutrol® dispersions: a thermal, microscopic and NMR relaxometry study. Pharm. Res.
F127 using hot melt extrusion. In: 39th CRS Annual Meeting & Exposition. Centre 27, 1869–1883.
des Congrès de Québec, Québec City, Canada (abstract ID: 100626). Qi, S., Belton, P., Nollenberger, K., Gryczke, A., Craig, D.Q., 2011. Compositional analy-
Ma, D., Djemai, A., Gendron, C.M., Xi, H., Smith, M., Kogan, J., Li, L., 2012. Development sis of low quantities of phase separation in hot-melt-extruded solid dispersions:
of a HPMC-based controlled release formulation with hot melt extrusion (HME). a combined atomic force microscopy, photothermal fourier-transform infrared
Drug Dev. Ind. Pharm, http://dx.doi.org/10.3109/03639045.2012.702350. microspectroscopy, and localised thermal analysis approach. Pharm. Res. 28,
Mackin, L., Zanon, R., Park, J.M., Foster, K., Opalenik, H., Demonte, M., 2002. Quantifi- 2311–2326.
cation of low levels (<10%) of amorphous content in micronised active batches Qi, S., Craig, D.Q., 2010. Detection of phase separation in hot melt extruded solid
using dynamic vapour sorption and isothermal microcalorimetry. Int. J. Pharm. dispersion formulations global vs localized characterization. Am. Pharm. Rev.,
231, 227–236. September 01.
Maclean, J., Medina, C., Daurio, D., Alvarez-Nunez, F., Jona, J., Munson, E., Nagapudi, K., Qi, S., Gryczke, A., Belton, P., Craig, D.Q., 2008. Characterisation of solid disper-
2011. Manufacture and performance evaluation of a stable amorphous complex sions of paracetamol and EUDRAGIT E prepared by hot-melt extrusion using
of an acidic drug molecule and neusilin. J. Pharm. Sci. 100, 3332–3344. thermal, microthermal and spectroscopic analysis. Int. J. Pharm. 354, 158–
Marsac, P.J., Konno, H., Rumondor, A.C., Taylor, L.S., 2008. Recrystallization of nifedip- 167.
ine and felodipine from amorphous molecular level solid dispersions containing Qian, F., Huang, J., Hussain, M.A., 2010. Drug–polymer solubility and miscibility:
poly(vinylpyrrolidone) and sorbed water. Pharm. Res. 25, 647–656. stability consideration and practical challenges in amorphous solid dispersion
Marsac, P.J., Konno, H., Taylor, L.S., 2006. A comparison of the physical stability of development. J. Pharm. Sci. 99, 2941–2947.
amorphous felodipine and nifedipine systems. Pharm. Res. 23, 2306–2316. Ranzani, L.S., Font, J., Galimany, F., Santanach, A., Gomez-Gomar, A.M., Casadevall,
Marsac, P.J., Rumondor, A.C., Nivens, D.E., Kestur, U.S., Stanciu, L., Taylor, L.S., 2010. G., Gryczke, A., 2011. Enhanced in vivo absorption of CB-1 antagonist in rats via
Effect of temperature and moisture on the miscibility of amorphous dispersions solid solutions prepared by hot-melt extrusion. Drug Dev. Ind. Pharm. 37 (6),
of felodipine and poly(vinyl pyrrolidone). J. Pharm. Sci. 99, 169–185. 694–701.
Mathot, V.B.F., Goderis, B., Scherrenberg, R.L., van der Vegte, E.W., 2002. High-speed Rathore, A.S., Bhambure, R., Ghare, V., 2010. Process analytical technology (PAT) for
calorimetry for the study of the kinetics of (de)vitrification, crystallization, and biopharmaceutical products. Anal. Bioanal. Chem. 398, 137–154.
melting of macromolecules. Macromolecules 35, 3601–3613. Rauwendaal, C., 1994. Polymer Extrusion, 3rd ed. Hanser Publisher, Munich.
Matsumoto, T., Zografi, G., 1999. Physical properties of solid molecular dispersions of Rauwendaal, C., 2001. Polymer Extrusion, 3rd ed. Hanser Publisher, Munich.
indomethacin with poly(vinylpyrrolidone) and poly(vinylpyrrolidone-co-vinyl- Read, E.K., Park, J.T., Shah, R.B., Riley, B.S., Brorson, K.A., Rathore, A.S., 2010. Process
acetate) in relation to indomethacin crystallization. Pharm. Res. 16, 1722–1728. analytical technology (PAT) for biopharmaceutical products: Part I. concepts and
Mccrum, N.G., Buckley, C.P., 1997. Principles of Polymer Engineering. Oxford Uni- applications. Biotechnol. Bioeng. 105, 276–284.
versity Press, Oxford. Reitz, C., Strachan, C., Kleinebudde, P., 2008. Solid lipid extrudates as sustained-
McGinity, J.W., Koleng, J.J., 1997. Preparation and evaluation of rapid-release gran- release matrices: the effect of surface structure on drug release properties. Eur.
ules using a novel hot-melt extrusion technique. In: Pharmaceutical Technology J. Pharm. Sci. 35, 335–343.
Conference. Pharmaceutical Technology, pp. 153–154. Repka, M.A., Battu, S.K., Upadhye, S.B., Thumma, S., Crowley, M.M., Zhang, F., Martin,
Mididoddi, P.K., Repka, M.A., 2007. Characterization of hot-melt extruded drug deliv- C., McGinity, J.W., 2007. Pharmaceutical applications of hot-melt extrusion: part
ery systems for onychomycosis. Eur. J. Pharm. Biopharm. 66, 95–105. II. Drug Dev. Ind. Pharm. 33, 1043–1057.
Miller, D.A., DiNunzio, J.C., Yang, W., McGinity, J.W., Williams 3rd, R.O., 2008. Repka, M.A., Gerding, T.G., Repka, S.L., McGinity, J.W., 1999. Influence of plasticiz-
Targeted intestinal delivery of supersaturated itraconazole for improved oral ers and drugs on the physical–mechanical properties of hydroxypropylcellulose
absorption. Pharm. Res. 25, 1450–1459. films prepared by hot melt extrusion. Drug Dev. Ind. Pharm. 25, 625–633.
Miller, D.A., McConville, J.T., Yang, W., Williams 3rd, R.O., McGinity, J.W., 2007. Hot- Repka, M.A., Majumdar, S., Kumar Battu, S., Srirangam, R., Upadhye, S.B., 2008. Appli-
melt extrusion for enhanced delivery of drug particles. J. Pharm. Sci. 96, 361–376. cations of hot-melt extrusion for drug delivery. Expert Opin. Drug Deliv. 5,
Miyazaki, T., Yoshioka, S., Aso, Y., Kojima, S., 2004. Ability of polyvinylpyrrolidone 1357–1376.
and polyacrylic acid to inhibit the crystallization of amorphous acetaminophen. Repka, M.A., McGinity, J.W., Zhang, F., 2002. Hot-Melt Extrusion Technology, 2nd ed.
J. Pharm. Sci. 93, 2710–2717. Marcel Dekker Inc., New York.
Mohammed, N., Majumdar, S., Repka, M.A., Durig, T.,2011. Tablets manufactured via Repka, M.A., Shah, S., Lu, J., Maddineni, S., Morott, J., Patwardhan, K., Mohammed,
HME as a processing technique for solubility enhancement: stability assessment N.N., 2012. Melt extrusion: process to product. Expert Opin. Drug Deliv. 9,
and in vitro release comparison to a marketed fromulation. In: American Associ- 105–125.
ation of Pharmaceutical Scientists Annual Meeting and Exposition. Washington Roblegg, E., Jager, E., Hodzic, A., Koscher, G., Mohr, S., Zimmer, A., Khinast, J., 2011.
Convention Center, Washington, DC, USA (abstract ID: #T3277). Development of sustained-release lipophilic calcium stearate pellets via hot
Moran, A., Buckton, G., 2009. Studies of the crystallization of amorphous trehalose melt extrusion. Eur. J. Pharm. Biopharm. 79, 635–645.
using simultaneous gravimetric vapor sorption/near IR (GVS/NIR) and modu- Rodrigues, L.O., Alves, T.P., Cardoso, J.P., Menezes, J.C., 2006. Improving drug manu-
lated GVS/NIR. AAPS PharmSciTech 10, 297–302. facturing with process analytical technology. IDrugs 9, 44–48.
Nakamichi, K., Nakano, T., Yasuura, H., Izumi, S., Kawashima, Y., 2002. The role of the Rumondor, A., Ivanisevic, I., Bates, S., Alonzo, D., Taylor, L., 2009a. Evaluation of
kneading paddle and the effects of screw revolution speed and water content on drug–polymer miscibility in amorphous solid dispersion systems. Pharm. Res.
the preparation of solid dispersions using a twin-screw extruder. Int. J. Pharm. 26, 2523–2534.
241, 203–211. Rumondor, A.C., Marsac, P.J., Stanford, L.A., Taylor, L.S., 2009b. Phase behavior of
Newman, A., Engers, D., Bates, S., Ivanisevic, I., Kelly, R.C., Zografi, G., 2008. Charac- poly(vinylpyrrolidone) containing amorphous solid dispersions in the presence
terization of amorphous API:polymer mixtures using X-ray powder diffraction. of moisture. Mol. Pharm. 6, 1492–1505.
J. Pharm. Sci. 97, 4840–4856. Rumondor, A.C., Taylor, L.S., 2010. Application of partial least-squares (PLS) model-
Newman, A., Knipp, G., Zografi, G., 2012. Assessing the performance of amorphous ing in quantifying drug crystallinity in amorphous solid dispersions. Int. J. Pharm.
solid dispersions. J. Pharm. Sci. 101, 1355–1377. 398, 155–160.
Nishi, T., Wang, T.T., 1975. Melting point depression and kinetic effects of cool- Rumondor, A.C., Wikstrom, H., Van Eerdenbrugh, B., Taylor, L.S., 2011.
ing on crystallization in poly(vinylidene fluoride)–poly(methyl methacrylate) Understanding the tendency of amorphous solid dispersions to undergo
mixtures. Macromolecules 8, 909–915. amorphous–amorphous phase separation in the presence of absorbed
Nollenberger, K., Gryczke, A., Meier, C., Dressman, J., Schmidt, M.U., Bruhne, S., 2009. moisture. AAPS PharmSciTech 12, 1209–1219.
Pair distribution function X-ray analysis explains dissolution characteristics of Saerens, L., Dierickx, L., Lenain, B., Vervaet, C., Remon, J.P., De Beer, T., 2011. Raman
felodipine melt extrusion products. J. Pharm. Sci. 98, 1476–1486. spectroscopy for the in-line polymer–drug quantification and solid state char-
O’Neill, M.A., Gaisford, S., 2011. Application and use of isothermal calorimetry in acterization during a pharmaceutical hot-melt extrusion process. Eur. J. Pharm.
pharmaceutical development. Int. J. Pharm. 417, 83–93. Biopharm. 77, 158–163.
Onoue, S., Takahashi, H., Kawabata, Y., Seto, Y., Hatanaka, J., Timmermann, B., Saerens, L., Dierickx, L., Quinten, T., Adriaensens, P., Carleer, R., Vervaet, C., Remon,
Yamada, S., 2010. Formulation design and photochemical studies on nanocrys- J.P., De Beer, T., 2012. In-line NIR spectroscopy for the understanding of
tal solid dispersion of curcumin with improved oral bioavailability. J. Pharm. Sci. polymer–drug interaction during pharmaceutical hot-melt extrusion. Eur. J.
99, 1871–1881. Pharm. Biopharm. 81 (1), 230–237.
252 S. Shah et al. / International Journal of Pharmaceutics 453 (2013) 233–252

Sakurai, A., Sakai, T., Sako, K., Maitani, Y., 2012. Polymer combination increased Tumuluri, V.S., Kemper, M.S., Lewis, I.R., Prodduturi, S., Majumdar, S., Avery, B.A.,
both physical stability and oral absorption of solid dispersions containing a low Repka, M.A., 2008. Off-line and on-line measurements of drug-loaded hot-melt
glass transition temperature drug: physicochemical characterization and in vivo extruded films using Raman spectroscopy. Int. J. Pharm. 357, 77–84.
study. Chem. Pharm. Bull. (Tokyo) 60, 459–464. Upadhye, S.B., Kulkarni, S.J., Majumdar, S., Avery, M.A., Gul, W., ElSohly, M.A.,
Saleki-Gerhardt, A., Ahlneck, C., Zografi, G., 1994. Assessment of disorder in crys- Repka, M.A., 2010. Preparation and characterization of inclusion complexes of
talline solids. Int. J. Pharm. 101, 237–247. a hemisuccinate ester prodrug of delta9-tetrahydrocannabinol with modified
Sathigari, S.K., Radhakrishnan, V.K., Davis, V.A., Parsons, D.L., Babu, R.J., 2012. beta-cyclodextrins. AAPS PharmSciTech 11, 509–517.
Amorphous-state characterization of efavirenz-polymer hot-melt extrusion Uzun, I.N., Sipahigil, O., Dincer, S., 2011. Coprecipitation of Cefuroxime Axetil-PVP
systems for dissolution enhancement. J. Pharm. Sci. 101 (9), 3456–3464. composite microparticles by batch supercritical antisolvent process. The Journal
Saunders, M., Podluii, K., Shergill, S., Buckton, G., Royall, P., 2004. The potential of high of Supercritical Fluids 55, 1059–1069.
speed DSC (hyper-DSC) for the detection and quantification of small amounts Van den Mooter, G., 2012. The use of amorphous solid dispersions: a formulation
of amorphous content in predominantly crystalline samples. Int. J. Pharm. 274, strategy to overcome poor solubility and dissolution rate. Drug Discov. Today:
35–40. Technol. 9, e79–e85.
Schilling, S.U., Bruce, C.D., Shah, N.H., Malick, A.W., McGinity, J.W., 2008. Citric acid Van den Mooter, G., Wuyts, M., Blaton, N., Busson, R., Grobet, P., Augustijns, P., Kinget,
monohydrate as a release-modifying agent in melt extruded matrix tablets. Int. R., 2001. Physical stabilisation of amorphous ketoconazole in solid dispersions
J. Pharm. 361, 158–168. with polyvinylpyrrolidone K25. Eur. J. Pharm. Sci. 12, 261–269.
Schilling, S.U., Lirola, H.L., Shah, N.H., Waseem Malick, A., McGinity, J.W., 2010. Influ- Van Eerdenbrugh, B., Lo, M., Kjoller, K., Marcott, C., Taylor, L.S., 2012a. Nanoscale
ence of plasticizer type and level on the properties of Eudragit S100 matrix mid-infrared evaluation of the miscibility behavior of blends of dextran or mal-
pellets prepared by hot-melt extrusion. J. Microencapsul. 27, 521–532. todextrin with poly(vinylpyrrolidone). Mol. Pharm. 9, 1459–1469.
Schilling, S.U., Shah, N.H., Malick, A.W., Infeld, M.H., McGinity, J.W., 2007. Citric Van Eerdenbrugh, B., Lo, M., Kjoller, K., Marcott, C., Taylor, L.S., 2012b. Nanoscale
acid as a solid-state plasticizer for Eudragit RS PO. J. Pharm. Pharmacol. 59, mid-infrared imaging of phase separation in a drug–polymer blend. J. Pharm.
1493–1500. Sci. 101, 2066–2073.
Scott, B., Wilcock, A., 2006. Process analytical technology in the pharmaceutical Vasconcelos, T., Sarmento, B., Costa, P., 2007. Solid dispersions as strategy to
industry: a toolkit for continuous improvement. PDA J. Pharm. Sci. Technol. 60, improve oral bioavailability of poor water soluble drugs. Drug Discov. Today
17–53. 12, 1068–1075.
Serajuddin, A.T., 1999. Solid dispersion of poorly water-soluble drugs: early Verhoeven, E., De Beer, T.R., Van den Mooter, G., Remon, J.P., Vervaet, C., 2008. Influ-
promises, subsequent problems, and recent breakthroughs. J. Pharm. Sci. 88, ence of formulation and process parameters on the release characteristics of
1058–1066. ethylcellulose sustained-release mini-matrices produced by hot-melt extrusion.
Shah, N., Choi, S.H.D.S., Kalb, O., Page, S., Wyttenbach, N., 2012. Structured develop- Eur. J. Pharm. Biopharm. 69, 312–319.
ment approach for amorphous systems. In: Williams III, R.O., Watts, A.B., Miller, Verhoeven, E., De Beer, T.R.M., Schacht, E., Van den Mooter, G., Remon, J.P., Vervaet,
D.A. (Eds.), Formulating Poorly Water Soluble Drugs. Springer, New York, NY, C., 2009. Influence of polyethylene glycol/polyethylene oxide on the release
pp. 267–310. characteristics of sustained-release ethylcellulose mini-matrices produced by
Sherry, R., 2007. Reckitt Benckiser Healthcare (UK) Limited Assignee. Granules com- hot-melt extrusion: in vitro and in vivo evaluations. Eur. J. Pharm. Biopharm.
prising paracetamol, a NSAID and a sugar alcohol made by melt extrusion patent. 72, 463–470.
Shibata, Y., Fujii, M., Sugamura, Y., Yoshikawa, R., Fujimoto, S., Nakanishi, S., Moto- Verreck, G., Six, K., Van den Mooter, G., Baert, L., Peeters, J., Brewster, M.E., 2003.
sugi, Y., Koizumi, N., Yamada, M., Ouchi, K., Watanabe, Y., 2009. The preparation Characterization of solid dispersions of itraconazole and hydroxypropylmethyl-
of a solid dispersion powder of indomethacin with crospovidone using a twin- cellulose prepared by melt extrusion—part I. Int. J. Pharm. 251, 165–174.
screw extruder or kneader. Int. J. Pharm. 365, 53–60. Vyazovkin, S., Dranca, I., 2007. Effect of physical aging on nucleation of amorphous
Six, K., Murphy, J., Weuts, I., Craig, D.Q., Verreck, G., Peeters, J., Brewster, M., Van indomethacin. J. Phys. Chem. B 111, 7283–7287.
den Mooter, G., 2003. Identification of phase separation in solid dispersions of Yang, M., Wang, P., Gogos, C., 2012. Prediction of acetaminophen’s solubility in
itraconazole and Eudragit E100 using microthermal analysis. Pharm. Res. 20, poly(ethylene oxide) at room temperature using the Flory-Huggins theory. Drug
135–138. Dev. Ind. Pharm., http://dx.doi.org/10.3109/03639045.2012.659188.
Smith, A.J., Kavuru, P., Wojtas, L., Zaworotko, M.J., Shytle, R.D., 2011. Cocrystals Yang, M., Wang, P., Suwardie, H., Gogos, C., 2011. Determination of acetaminophen’s
of quercetin with improved solubility and oral bioavailability. Mol. Pharm. 8, solubility in poly(ethylene oxide) by rheological, thermal and microscopic meth-
1867–1876. ods. Int. J. Pharm. 403, 83–89.
Steiner, R., 2003. Extruder design. In: Ghebre-Sellassie, I.M.C. (Ed.), Pharmaceutical Yano, H., Kleinebudde, P., 2010. Improvement of dissolution behavior for poorly
Extrusion Technology. Marcel Dekker, New York, pp. 20–38. water-soluble drug by application of cyclodextrin in extrusion process:
Sun, Y., Rui, Y., Wenliang, Z., Tang, X., 2008. Nimodipine semi-solid capsules con- comparison between melt extrusion and wet extrusion. AAPS PharmSciTech 11,
taining solid dispersion for improving dissolution. Int. J. Pharm. 359, 144–149. 885–893.
Taylor, L.S., Zografi, G., 1997. Spectroscopic characterization of interactions between Yoo, S.U., Krill, S.L., Wang, Z., Telang, C., 2009. Miscibility/stability considerations in
PVP and indomethacin in amorphous molecular dispersions. Pharm. Res. 14, binary solid dispersion systems composed of functional excipients towards the
1691–1698. design of multi-component amorphous systems. J. Pharm. Sci. 98, 4711–4723.
Thiele, W., 2003. Twin-screw extrusion and screw design. In: Ghebre-Sellassie, Yoshioka, M., Hancock, B.C., Zografi, G., 1995. Inhibition of indomethacin crystalliza-
I.M.C. (Ed.), Pharmaceutical Extrusion Technology. Marcel Dekker, New York, tion in poly(vinylpyrrolidone) coprecipitates. J. Pharm. Sci. 84, 983–986.
pp. 69–98. Yu, L.X., Lionberger, R.A., Raw, A.S., D’Costa, R., Wu, H., Hussain, A.S., 2004. Appli-
Tho, I., Liepold, B., Rosenberg, J., Maegerlein, M., Brandl, M., Fricker, G., 2010. For- cations of process analytical technology to crystallization processes. Adv. Drug
mation of nano/micro-dispersions with improved dissolution properties upon Deliv. Rev. 56, 349–369.
dispersion of ritonavir melt extrudate in aqueous media. Eur. J. Pharm. Sci. 40, Zhao, Y., Inbar, P., Chokshi, H.P., Malick, A.W., Choi, D.S., 2011. Prediction of the ther-
25–32. mal phase diagram of amorphous solid dispersions by Flory–Huggins theory.
Thommes, M., Baert, L., Rosier, J., 2010. 800 mg Darunavir tablets prepared by hot J. Pharm. Sci. 100, 3196–3207.
melt extrusion. Pharm. Dev. Technol. 16 (6), 645–650. Zheng, X., Yang, R., Tang, X., Zheng, L., 2007. Part I: characterization of solid disper-
Thommes, M., Ely, D.R., Carvajal, M.T., Pinal, R., 2011. Improvement of the dissolution sions of nimodipine prepared by hot-melt extrusion. Drug Dev. Ind. Pharm. 33,
rate of poorly soluble drugs by solid crystal suspensions. Mol. Pharm. 8, 727–735. 791–802.

You might also like