You are on page 1of 25

Journal of Controlled Release 248 (2017) 7195

Contents lists available at ScienceDirect

Journal of Controlled Release

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

Review article

BCS class IV drugs: Highly notorious candidates for


formulation development
Rohan Ghadi a,, Neha Dand b
a
IPDO, Innovation Plaza, Dr Reddy's Laboratories Ltd., Bachupally, Hyderabad, 500090, India
b
Department of Pharmaceutics, Bharati Vidyapeeth's College of Pharmacy, CBD Belapur, Navi Mumbai, 400064, India

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

Article history: BCS class IV drugs (e.g., amphotericin B, furosemide, acetazolamide, ritonavir, paclitaxel) exhibit many character-
Received 14 October 2016 istics that are problematic for effective oral and per oral delivery. Some of the problems associated include low
Accepted 8 January 2017 aqueous solubility, poor permeability, erratic and poor absorption, inter and intra subject variability and signi-
Available online 11 January 2017
cant positive food effect which leads to low and variable bioavailability. Also, most of the class IV drugs are sub-
strate for P-glycoprotein (low permeability) and substrate for CYP3A4 (extensive pre systemic metabolism)
Keywords:
Bioavailability
which further potentiates the problem of poor therapeutic potential of these drugs. A decade back, extreme ex-
Dissolution rate amples of class IV compounds were an exception rather than the rule, yet today many drug candidates under de-
Enhanced permeation velopment pipeline fall into this category. Formulation and development of an efcacious delivery system for BCS
P-gp efux class IV drugs are herculean tasks for any formulator. The inherent hurdles posed by these drugs hamper their
translation to actual market. The importance of the formulation composition and design to successful drug devel-
opment is especially illustrated by the BCS class IV case. To be clinically effective these drugs require the devel-
opment of a proper delivery system for both oral and per oral delivery. Ideal oral dosage forms should produce
both a reasonably high bioavailability and low inter and intra subject variability in absorption. Also, ideal systems
for BCS class IV should produce a therapeutic concentration of the drug at reasonable dose volumes for intrave-
nous administration. This article highlights the various techniques and upcoming strategies which can be
employed for the development of highly notorious BCS class IV drugs. Some of the techniques employed are
lipid based delivery systems, polymer based nanocarriers, crystal engineering (nanocrystals and co-crystals),
liquisolid technology, self-emulsifying solid dispersions and miscellaneous techniques addressing the P-gp efux
problem. The review also focuses on the roadblocks in the clinical development of the aforementioned strategies
such as problems in scale up, manufacturing under cGMP guidelines, appropriate quality control tests, validation
of various processes and variable therein etc. It also brings to forefront the current lack of regulatory guidelines
which poses difculties during preclinical and clinical testing for submission of NDA and subsequent marketing.
Today, the pharmaceutical industry has as its disposal a series of reliable and scalable formulation strategies for
BCS Class IV drugs. However, due to lack of understanding of the basic physical chemistry behind these strategies
formulation development is still driven by trial and error.
2017 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 72
2. Lipid based drug delivery systems (LBDDS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
2.1. Mechanism of improved bioavailability of BCS class IV drugs by LBDDS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 73
2.2. Potential applicability of LBDDS for improving bioavailability of BCS class IV drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2.2.1. Emulsions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2.2.2. Self-emulsifying drug delivery systems (SEDDS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2.2.3. Liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
2.2.4. Solid lipid nanoparticles (SLNs) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77

Corresponding author.
E-mail address: rohanghadi13@gmail.com (R. Ghadi).

http://dx.doi.org/10.1016/j.jconrel.2017.01.014
0168-3659/ 2017 Elsevier B.V. All rights reserved.
72 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

2.2.5. Nanostructured lipid carriers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77


2.2.6. Lipid polymer hybrid nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
2.2.7. Lipid nanocapsules . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3. Polymer nanocarrier based approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 78
3.1. Mechanism of improved bioavailability of BCS class IV drugs by polymer nanocarrier based approach. . . . . . . . . . . . . . . . . . . . 78
3.2. Potential applicability of polymer based nanocarriers for improving the bioavailability of BCS class IV drugs . . . . . . . . . . . . . . . . . 79
3.2.1. Polymeric micelles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
3.2.2. Polymeric nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
3.2.3. Dendrimers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4. Pharmaceutical crystal engineering . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.1. Nanocrystal technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 79
4.1.1. Mechanism of improved bioavailability by nanocrystal technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
4.1.2. Potential applicability of nanocrystal technology to BCS class IV drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 80
4.2. Co-crystal technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
4.2.1. Potential applicability of co-crystal technology to BCS class IV drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5. Liquisolid technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.1. Mechanism of improved bioavailability of BCS class IV drugs by liquisolid technology . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.1.1. Increased drug surface area . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.1.2. Increased drug solubility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
5.1.3. Improved wetting properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
5.2. Potential applicability of liquisolid technology to BCS IV class drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
6. Self-emulsifying solid dispersion formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
6.1. Examples of surface active excipients used in the fabrication of self-emulsifying solid dispersions. . . . . . . . . . . . . . . . . . . . . . 84
6.1.1. Gelucires. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
6.1.2. Tocopheryl polyethylene glycol 1000 succinate (TPGS). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
6.1.3. Pluronics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
6.2. Mechanism of improved bioavailability of BCS class IV drugs by self-emulsifying solid dispersion formulations. . . . . . . . . . . . . . . . 84
6.3. Potential applicability of self-emulsifying solid dispersion formulations to BCS class IV drugs . . . . . . . . . . . . . . . . . . . . . . . . 85
7. P-glycoprotein (P-gp) inhibition strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
7.1. P-gp inhibition by lipidic and polymeric excipients and formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
7.2. P-gp inhibition by using specic and non-specic P-gp inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
7.3. P-gp circumvention by using novel peptide prodrug approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
8. Current roadblocks in the clinical development of formulation strategies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88
9. Regulatory aspects for the development of BCS class IV delivery systems . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89
10. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 92

1. Introduction low systemic exposure, result in adverse effects because of the high
dose and may vary in their effect in individual patients [7].
The rate and extent of drug absorption from the gastrointestinal (GI) Based on the BCS, drugs are classied into four categories according
tract are very intricate and affected by many factors. These include to their solubility and permeability properties as follows; high solubili-
physicochemical factors, physiological factors, and factors related to tyhigh permeability (class I); low solubilityhigh permeability (class
the dosage form [1,2]. Despite this complexity, the Biopharmaceutics II); high solubilitylow permeability (class III); and low solubilitylow
Classication System (BCS) developed by Amidon et al. [3] revealed permeability (class IV) [3]. The drugs exhibiting low solubility but rea-
that the essential key parameters controlling oral drug absorption are sonable membrane permeability such as phenytoin, glibenclamide, car-
the solubility/dissolution of the drug dose in the GI milieu and the per- bamazepine, ibuprofen etc. are categorised as BCS class II. The rate-
meability of the drug through the GI membrane. These important pa- limiting process of absorption for class II drugs is the dissolution step.
rameters are characterized in the BCS as one of the most signicant Formulation plays a major role in determining the rate and extent of ab-
tools in modern pharmaceutics and biopharmaceutics of oral drug prod- sorption of such drugs from the gastrointestinal tract. A number of for-
ucts. The progress made in combinatorial chemistry and innovative mulation strategies have been developed to improve the delivery of BCS
high-throughput screening has led to the production of a vast number class II drugs like complexation, micronization, crystal modication etc.
of potential drug candidates. However, at the same time these tech- They are based on techniques either to increase the drug dissolution
niques have introduced more poorly water-soluble drugs in the phar- rate or to achieve sustained solubilization of the drugs. The poorly
maceutical pipeline. It is estimated that N40% of marketed drugs are water-soluble drugs with poor membrane permeability (Table 1) be-
poorly water-soluble [4,5]. Lipinski et al. pointed out that leads obtained long to BCS class IV such as amphotericin B (AmB), furosemide (FUR),
through high-throughput screening (HTS) tend to have higher molecu- acetazolamide, ritonavir (RTV) etc. Usually techniques used for BCS
lar weights and greater lipophilicity than leads in the pre-HTS era [4]. class II drugs do little to improve the absorption of class IV drugs due
Dissolution of the drug in the aqueous milieu of the GI is almost always to the limited membrane permeability. As a result, the best solution to
a prerequisite for oral absorption, and hence, inadequate aqueous solu- improve the bioavailability of class IV drugs is to go back to the lead op-
bility often causes limited or poor oral bioavailability. Furthermore the timisation phase of drug discovery and modify their structures to obtain
problems associated with poor stability and membrane permeability the appropriate physicochemical properties. However, discovering a
adds on to the dilemma of low bioavailability [4,6]. All these problems novel therapeutic agent, itself is a challenging, time-consuming and
are plaguing the lead molecules or drug candidates in the pipeline of costly process. It takes US $8001200 million and 1015 years to devel-
most major pharmaceutical companies. Low/variable bioavailability of op a new chemical entity. Also, very few of the millions of compounds
a drug is one of the major hurdles for formulation scientists, because it after being tested reach the market. Hence, sending a drug molecule
can lead to compromised product performance and the drug is unlikely back to the lead optimisation phase is not a feasible option because of
to reach its molecular target. Such drugs require a high dose but have the constraints associated with time, cost, labour and resources. As a
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 73

Table 1
List of BCS Class IV drugs according to the WHO model list of essential medicines and reported literature.

Drug Category BCS class References

Drugs with reliable solubility and permeability data Aluminium hydroxide Gastro intestinal agent Class IV [8]
(WHO model list of Essential medicines) Furosemide (FUR) Diuretic Class IVa,b [813]
Indinavir (IDV) Anti-viral Class IVa,c,d [8]
Nelnavir (NFV) Anti-viral Class IVa,c,d [8]
Ritonavir (RTV) Anti-viral Class IVa,c,d [8,14]
Saquinavir (SQV) Anti-viral Class IVa,c,d [8,14,15]
Acetazolamide Diuretic Class IVb [8,16]
Drugs for which complete solubility and/or permeability data are lacking data Azathioprine Immunosuppressive Class IV [8,16]
(WHO model list of Essential medicines) Amphotericin B (AmB) Anti-fungal Class IV [1719]
Drugs with solubility and permeability issues (Reported in literature) Hydrochlorothiazide (HCT) Diuretic Class IV [19,20]
Colistin Anti-microbial Class IV [19]
Chlorthalidone Anti-hypertensive Class IV [19]
Chlorothiazide Diuretic Class IV [19]
Cyclosporine A (CyA) Immunosuppressive Class IVc [14,21]
Paclitaxel (PTX) Anti-cancer Class IVa,d [2225]
Docetaxel (DTX) Anti-cancer Class IVa,d [25]
Famotidine (FAM) Gastro protective agent Class IVb [11,26]
Aprepitant Anti-emetic Class IV [14]
Lopinavir (LPV) Anti-viral Class IVa,c,d [27]
Etravirine Anti-viral Class IVa,d,c [28]
a
First pass effect.
b
Substrate for uptake transporter.
c
Signicant positive food effect.
d
Substrate for CYP3A4.

result, proper formulation is of key importance to establish a successful and tailored made oils and lipids have reached the market to aid in
product for the administration of BCS class IV drugs. Fig. 1. gives an in- easy formulation development.
sight on the various BCS IV drugs and the numerous hurdles posed by
them in the successful oral and peroral delivery. This article highlights 2.1. Mechanism of improved bioavailability of BCS class IV drugs by LBDDS
the various strategies which can be employed for improving the bio-
availability and successful delivery of BCS class IV drugs. These strate- The balance between a drug's solubility in the aqueous environment
gies include lipid based delivery systems, polymeric nanoparticulate of the gastrointestinal lumen and its permeation across the lipophilic
systems, crystal engineering (nanocrystal technology, co-crystal tech- membrane of enterocytes determines its rate and extent of absorption.
nology), liquisolid technology, self-emulsifying solid dispersions and After oral administration of lipid-based formulations, gastric lipase initi-
P-efux inhibition strategies. ates the digestion of exogenous dietary triglyceride (TG) and formula-
tion TG. Simultaneously, the mechanical mixing (propulsion, grinding
and retropulsion) of the stomach facilitates formation of a crude emul-
2. Lipid based drug delivery systems (LBDDS) sion (comprised of aqueous gastric uid and lipid digestion products).
Later in the small intestine, TG is broken down to diglyceride, mono-
LBDDS have gained increasing interest in pharmaceutical eld be- glyceride and fatty acids by pancreatic lipase together with its cofactor
cause of their potential to improve the bioavailability of drug molecules co- lipase 203, acting primarily at the sn-1 and sn-3 positions of TG to
which are difcult to formulate [29]. There is a current resurgence of in- produce 2-monoglyceride and free fatty acid. Pancreatic phospholipase
terest in lipid based dosage forms because of the proven commercial A2 digests the formulation-derived or biliary-derived phospholipids
and pharmaceutical benet for compounds such as CyA, lipid soluble vi- (PL) by hydrolyzing at the sn-2 position of PL to yield
tamins, and the HIV protease inhibitors. Also, the industry is trending lysophosphatidylcholine and fatty acid. The presence of exogenous
toward the discovery/development of increasingly hydrophobic (and lipids in the small intestine stimulates the secretion of endogenous
potent) new chemical entities. Finally, the resolution of many of the pre- biliary lipids from the gall bladder, including bile salt (BS), PL and
viously problematic technology transfer, stability and regulatory issues cholesterol. Previously formed monoglycerides, fatty acids, and
makes lipid based delivery systems as a good option for clinical transla- lysophospholipid (products of lipid digestion) are subsequently incor-
tion of drug products. porated in to a series of colloidal structures, including micelles and
Lipids as carriers have the potential of providing endless opportuni- unilamellar and multi-lamellar vesicles in the presence of bile salts.
ties in the area of drug delivery due to their ability to enhance gastroin- The solubilization and absorptive capacity of the small intestine for
testinal solubilization and absorption by selective lymphatic uptake of lipid digestion products and drugs (D) is signicantly enhanced due to
poorly bioavailable drugs [30]. The different properties of lipids such these formed lipid metabolites.
as physiochemical diversity, biocompatibility etc. have an ability to en- The formed micelles will then be absorbed by enterocytes, where it
hance oral bioavailability of poorly water soluble, lipophilic drugs and gets converted to the chylomicrons upon re-esterication via monoacyl
also poorly permeable drugs like BCS class IV as shown in Fig. 2. This glycerol or phosphatidic acid pathway and subsequent stabilization by
could be attributed to some unique properties of lipids such as high sol- phospholipids. However, the penetration of unstirred water layer and
ubilization potential, manufacturing scalability, industrial adaptability, mucin in gastrointestinal tract is rate limiting factor. The formed chylo-
biocompatibility and distinct route of absorption thereby eliminating microns are then subjected to lymphatic transport system via mesenter-
various physiological barriers such as pre-systemic metabolism, gastro- ic lymph and ultimately enter the systemic circulation by lymphatic
intestinal degradation, P-gp efux and permeability related issues. The drainage at thoracic duct [31]. The other transcellular routes by which
lipid excipients mainly comprise of monoglycerides, diglycerides, tri- lipid based drug delivery systems get transported across enterocytes in-
glycerides, oils constituting various combinations of glycerides, phos- clude macropinocytosis, clathrin-mediated, caveolae-mediated, and
pholipids, sphingolipids and even high fat meal. Also, many modied clathrin- and caveolae-independent endocytosis (principally lipid rafts
74 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

Fig. 1. BCS Class IV drugs- the inherent hurdles posed by them in their oral and per oral delivery along with the strategies to overcome them.
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 75

Fig. 2. Key advantages associated with lipid based drug delivery systems which improve oral and peroral delivery of BCS class IV drugs.

comprising of sphingolipids- and cholesterol-rich microdomains) [32]. to improve drug absorption through mitigation of presystemic drug me-
Fig. 3. depicts various absorption mechanisms by which lipid tabolism associated with gut membrane-bound cytochrome P-450
nanocarriers improve the oral bioavailability of BCS Class IV drugs. enzymes or via inhibition of the P-glycoprotein efux transporter [33
More recently, certain lipids and lipidic excipients have been suggested 36]. Various types of lipid based nanocarriers implemented for

Fig. 3. Absorption mechanisms implemented by lipidic nanocarriers for improving the bioavailability of BCS class IV drugs.
76 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

improving the bioavailability of BCS class IV drugs include however it has been postulated that it happens when entropy change
microemulsions, nanoemulsions, lipid nanocapsules, self-emulsifying for dispersion exceeds the energy required to increase the surface area
systems, lipid nanoparticles, hybrid lipid nanoparticles, liposomes and of dispersion [42,43].
surface engineered liposomes to name a few. Recently, co-administration of DTX with curcumin loaded SEDDS led
to about 3.2-fold increase in the oral bioavailability of DTX as compared
2.2. Potential applicability of LBDDS for improving bioavailability of BCS to the free drug. The inhibitory effect of curcumin on P-gp and cyto-
class IV drugs chrome P450 was also attributed to this increment in oral bioavailability
of DTX [44]. Sultan et al. [45] investigated the potential of self-micro
Since many decades, lipids have been widely used for improving the emulsifying drug delivery systems (SMEDDS) and niosomes as carriers
oral bioavailability of various difcult-to-deliver drugs. The usual prob- for widening the gastrointestinal absorption window of FUR. The drug
lem associated with most of the drugs includes either low aqueous sol- was incorporated in SMEDDS and was encapsulated into niosomes.
ubility or poor intestinal permeability, both of which can adequately be The intestinal absorption was monitored at two anatomical sites (duo-
taken care by LBDDS [30]. Lipid-based formulations have emerged as an denum and jejuno-ileum) by employing in situ rabbit intestinal perfu-
effective and versatile technology for many BCS class IV drugs. The prin- sion technique. Perfusion of drug solution (control) revealed poor
cipal lipidic drug delivery systems and their application to improve the intestinal permeability with percent fraction absorbed (%Fa) from the
bioavailability of BCS Class IV drugs are highlighted in this section. duodenum and jejuno-ileum being 1.3 and 0.6% per cm, respectively.
Formulation of furosemide as SMEDDS increased the %Fa from the duo-
2.2.1. Emulsions denum and jejuno-ileum to reach 1.7 and 1% per cm, respectively.
Emulsion based approach has been one of the most ancient methods Niosomal encapsulation increased the %Fa from duodenum and
for delivery of lipophilic drugs. Presently two versions of emulsions viz. jejuno-ileum to record 1.9 and 1.2% per cm, respectively. The increase
microemulsions and nanoemulsions have been employed for improving in the %Fa was also revealed as a reduction in the length required for
the oral deliverability of BCS class IV drugs. The unique characteristics 95% absorption of the drug which was reduced from 557.2 to
include thermodynamic (microemulsions) or kinetic (nanoemulsions) 245.8 cm and to 279.8 cm after delivery as niosomes or SMEDDS, re-
stability, supersolvency, small droplet size, high industrial scalability spectively, in case of jejuno-ileum. The same trend was recorded with
(as low energy requirements for manufacturing) and use of lipidic ex- the duodenum.
cipients as absorption enhancers. Traditionally, these systems comprise Seo et al. prepared DTX loaded self-nano emulsifying drug delivery
of appropriate blend of oil, surfactant and co-surfactant dispersed in systems (SNEDDS) to improve the bioavailability and anti-tumour po-
aqueous phase tailor made as per the physicochemical properties of tential of DTX [46]. DTX-SNEDDS remarkably improved the systemic
drug substances [37]. circulation of DTX by augmenting the rate and extent of drug absorption
The oral administration of a microemulsion formulation containing and thereby bioavailability. However, the most important aspect of the
dimethyl isosorbide, Tween 80 and D, L--tocopherol and incorporating work was that Taxotere-induced toxicity was overcome by DTX-
PTX in the presence of P-gp inhibitor (KR-30032) to rats allowed PTX SNEDDS with a plausible anti-tumour response. Thus, DTX-SNEDDS of-
oral bioavailability to be increased from 4.6% to 41%. This effect was fered an exciting mode and opened a new arena for DTX delivery to im-
dose-dependent and was saturable above 20 mg/kg of KR-30032 [38]. prove its chemotherapeutic potential. Also, Chen et al. prepared a solid
Yin et al. prepared a microemulsion system of DTX and evaluated for supersaturable self-emulsifying drug delivery system (S-sSEDDS)
its solubilization capacity and oral bioavailability improvement [39]. using DTX [47]. The in vivo studies indicated that the area under the con-
Compared to the commercial product Taxotere, the apical to centrationtime curve (AUC0) of the DTX-S-sSEDDS increased by
basolateral (A B) transport of DTX across the Caco-2 cell monolayer nearly 8.77-fold, 1.45-fold more than those of the DTX powder and
from the microemulsion system was signicantly improved (0.624 g/ the conventional SEDDS. Similarly, Gao et al. explored PTX loaded (S-
cm2, p b 0.01). Moreover, the oral bioavailability of the system in rats sSEDDS) for improving the oral bioavailability of PTX [48]. Pharmacoki-
rose dramatically (34.42%) compared to that of the orally administered netic studies conducted in rats showed that the bioavailability of PTX in
Taxotere (6.63%). This increase in bioavailability was probably due to PTX-S-sSEDDS was 9.5% compared to 2% in the orally dosed Cremophor-
the combined effect of the enhancement in solubility, the inhibition of ethanol, a further increase in bioavailability to 22.6% was noted in the
P-gp efux system and the increase in permeability. These results en- presence of CyA. SMEDDS composed of -tocopherol, TPGS, propylene
couraged further development of DTX microemulsions as an oral drug glycol, sodium deoxycholate and Cremophor RH40 was used to im-
delivery system. Bhutani et al. investigated the potential of AmB prove the oral bioavailability of PTX. Compared with Taxol the oral
microemulsion for the treatment of invasive fungal disease [40]. The op- bioavailability of PTX included in SMEDDS increased by 28.652.7% at
timized microemulsion exhibited 2-fold higher drug permeation as various doses [49]. Also, Deshmukh et al. enhanced the dissolution
compared to plain drug solution. It also showed higher skin deposition, and oral bioavailability of RTV using the Solid Self-Microemulsifying
lower skin irritation and better anti-fungal activity. The in vitro anti-fun- Drug Delivery System (S-SMEDDS). S-SMEDDS improved the plasma
gal activity in Trichophyton rubrum fungal species showed higher zone prole in terms of maximum plasma concentration (Cmax), and area
of inhibition. The results indicated that, microemulsion can be used as under curve (AUC024h), which is almost two-folds higher than the
a promising alternative for AmB therapy. Also, Jha et al. attempted oral aqueous suspension of RTV [50].
microemulsion formulation of famotidine for enhancing the bioavail- Garg et al. developed solid self-nanoemulsifying oily formula-
ability [41]. The developed microemulsion system improved the perme- tions (S-SNEOFs) for enhancing the systemic bioavailability of LPV
ability (93.43%) by increasing the lipophilicity due to the oil phase and and targeting the same to the sanctuary site, i.e., lymphatic system
also by destabilizing the membrane stability due the surfactants. for complete HIV inhibition [51]. The performance evaluation
through in situ single pass intestinal perfusion (SPIP) studies as-
2.2.2. Self-emulsifying drug delivery systems (SEDDS) cribed the signicant enhancement in absorptivity parameters of
SEDDS are the most advanced approach of emulsion based drug de- the SNEOFs vis--vis the pure drug. Also, chylomicron ow block
livery systems and relies on the physiological uids for the in situ forma- SPIP studies revealed lymphatic uptake of LPV from the SNEOFs.
tion of micro/nanoemulsion. This formulation strategy comprises of Overall, in vivo pharmacokinetic studies in rats revealed signicant
drug dissolved in oils and stabilized by surfactants and co-surfactants, improvement in the rate and extent of oral bioavailability of the
which upon exposure to the aqueous environment under gentle agita- SNEOFs compared to the pure drug. These studies further substanti-
tion leads to spontaneous formation of emulsion. Although the exact ated the intestinal lymphatic transport of LPV for the management of
mechanism of self-emulsication is yet to be understood and is unclear the sanctuary site HIV. In a nutshell, the SNEOFs offered a complete
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 77

and holistic solution for the management of the viral loads in the Compritol-based solid lipid nanoparticles with a poloxamer coating
lymph and blood. can be effectively absorbed through the lymphatic system, prolong the
circulation of drug in blood by acting as a reservoir and can effectively
2.2.3. Liposomes target the drug to the CNS due to the combined effect of lipophilicity
Liposomes are the most sophisticated class of lipid based drug and surface charge. The high biocompatibility, biodegradability and
delivery systems known for improving the deliverability of various dif- nontoxicity of compritol make the compritol-based solid lipid nanopar-
cult-to-deliver drugs. These are vesicular structures prepared from ticles an excellent carrier for enhanced CNS delivery of LPV.
phospholipids and are capable of accommodating both hydrophilic Punna Rao et al. prepared stearic acid-based LPV loaded SLNs using a
and lipophilic drug substances within them [52,53]. These are reported hybrid design and compared in vivo performance of optimized formula-
to have huge potential in improving the bioavailability of various drugs tion with marketed LPV/RTV co-formulation [59]. Optimized SLNs ex-
[13,54]. hibited nanometric size (223 nm) with high entrapment efciency
Rathore et al. attempted mannosylated liposomes bearing AmB for (83%). In vitro drug release study of SLNs showed biphasic sustained re-
effective management of visceral leishmaniasis [54]. The in vivo anti- lease behaviour. Signicant increase in oral bioavailability of LPV from
leishmanial activity performed on Leishmania donovaniinfected golden LPV SLNs (5 folds) and LPV/RTV co-formulation (3.7 folds) was ob-
hamster, revealed that AmB solution showed reduction in the parasite served as compared with free LPV. LPV SLNs showed better tissue distri-
load by 42.5 1.8%. The mannose-coupled liposomes showed a maxi- bution of LPV in HIV reservoirs than LPV/RTV co-formulation. In vitro
mum reduction in parasite load (i.e., 78.8 3.9%). The biodistribution studies demonstrated that SLNs provided metabolic protection to LPV
study clearly showed the higher uptake of mannosylated liposomes in and went endocytosis during absorption.
the liver and spleen and hence the active targeting to the reticulo-endo-
thelial system, which, in turn, would provide a direct attack of the drug 2.2.5. Nanostructured lipid carriers
to the site where the pathogen resides, rendering the other organs free Nanostructured lipid carriers (NLCs) are the advanced generation of
and safe from the toxic manifestations of the drug. Wu et al. synthesized SLNs overcoming the problems associated with the later such as limited
and characterized a liposomal formulation of PTX targeting the folate re- drug loading capacity, restructuring during storage and subsequently
ceptor (FR), which was designed to overcome vehicle toxicity associat- expulsion of drug during storage. The said advantages could be attribut-
ed with the traditional Cremophor EL-based formulation and to provide ed to higher solubility of drugs in oils as compared to solid lipids.
the added advantages of prolonged systemic circulation time and selec- Beloqui et al. evaluated the potential of NLCs as a tool to enhance the
tive targeting of the FR. FR-targeted liposomes containing PTX showed oral bioavailability of SQV [15]. SQV transport across Caco-2 monolayers
3.8-fold greater cytotoxicity compared to non-targeted control lipo- was enhanced up to 3.5-fold by NLCs compared to SQV suspension. The
somes in KB cells [55]. Vural et al. developed chitosan coated liposomal size and amount of surfactant in the NLCs inuenced SQV's permeabili-
carrier system for delivery of FUR to improve its biopharmaceutical ty, the transcytosis pathway and the efux of SQV by P-gp. SQV-NLCs
characteristics [13]. An eight-fold increase in FUR permeability was ob- circumvented P-gp efux and used both caveolae- and clathrin-mediat-
tained for coated liposomes when compared to uncoated formulations. ed transcytosis. By modifying critical physicochemical parameters of the
According to the cytotoxicity studies, chitosan coated liposome formu- NLC formulation, the P-gp drug efux and also the transcytosis mecha-
lations showed less cytotoxicity than uncoated ones. Chitosan coating nism of the nanoparticles was altered. The ndings were encouraging
increased the viability of the liposomes. The permeation enhancer effect and opened a new option for the delivery of class IV drugs and P-gp sub-
observed for the chitosan coated liposomes make them attractive candi- strates by the oral route and support further nanotechnology ap-
dates that could be effective to improve bioavailability of Class IV drugs proaches on this regard. Also, the same group evaluated the
after oral administration. mucopenetrating properties of dextranprotamine (DexProt) coating
on NLCs as per SQV permeability enhancement. In the Caco-2 mono-
2.2.4. Solid lipid nanoparticles (SLNs) layers, DexProtNLCs increased upto 9-fold SQV permeability in com-
SLNs are the alternative version of emulsions in which the liquid oil parison to uncoated nanoparticles. In the Caco-2/HT29-MTX
is replaced by solid lipids. Specic advantages include modulation of monolayers, DexProtNLCs presenting a surface charge close to neu-
drug release, increased drug stability, exclusion of organic solvents trality signicantly increased SQV permeability. Hence, DexProt com-
from the manufacturing process, manufacturing scalability and indus- plex coating can be a promising strategy to ensure successful
trial adaptability [56]. In the latest years SLN offered new perspectives nanoparticle mucus-penetration, and thus, an efcient nanoparticle
in the formulation of poorly water-soluble drugs. oral delivery [60].
In this context, Alex et al. [57] successfully encapsulated LPV in glyc-
eryl behenate based solid lipid nanoparticles (Lo-SLN) for its ultimate 2.2.6. Lipid polymer hybrid nanoparticles
use to target intestinal lymphatic vessels in combined To address the limitations of polymeric nanoparticles and liposomes,
chemotherapythe so-called Highly Active Anti-Retroviral Therapy a new generation delivery vehicle of therapeutics termed lipidpolymer
(HAART). From the intestinal lymphatic transport study, it became evi- hybrid nanoparticles (LPNs) has been developed. LPNs are coreshell
dent that SLN increased the cumulative percentage dose of LPV secreted nanoparticle structures comprising polymer cores and lipid/lipidPEG
into the lymph, which was 4.91-fold higher when compared with a con- shells, which exhibit complementary characteristics of both polymeric
ventional drug solution in methyl cellulose 0.5% (w/v) as suspending nanoparticles and liposomes, particularly in terms of their physical sta-
agent (Lo-MC). The percentage bioavailability was signicantly en- bility and biocompatibility. Signicantly, the LPNs have recently been
hanced. The AUC for the Lo-SLN was 2.13-fold higher than that obtained demonstrated to exhibit superior in vivo cellular delivery efcacy com-
for the Lo-MC of similar concentration. Negi et al. also developed glycer- pared to that obtained from polymeric nanoparticles and liposomes
yl behenate based SLNs of LPV to improve its oral bioavailability. Higher [61].
oral bioavailability (3.56-fold) was found for LPV loaded SLNs in com- Liu et al. developed folic acid conjugated nanoparticles of mixed lipid
parison to bulk LPV due to higher lymphatic drug transport (p b 0.05) monolayer shell and biodegradable polymer core (LPNP) for targeted
[27]. delivery of DTX [62]. After 0.5 h and 2 h incubation, the cellular uptake
Aji et al. developed SLNs to increase the availability of LPV in the CNS efciency of the non-targeted LPNPs was measured to be 18.99 0.75%
[58]. SLNs exhibited a Cmax and Tmax of 632.86 81.61 ng/mL and 25 and 25.39 0.54%, respectively. Instead, after 0.5 h and 2 h incubation,
7.75 min, respectively, with a signicant increase in bioavailability in a the cellular uptake efciency of the targeted LPNPs was measured to be
rat model compared with a free-drug suspension. An appreciable in- 26.24 0.68% and 39.09 0.64%, respectively. The targeting effect of
crease in cerebrospinal uid concentration was detected with SLNs. folic acid conjugation was thus signicant of 38.2% increment for 0.5 h
78 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

incubation and 54.0% increment for 2 h incubation, respectively. Also, transporter inhibition, targeting potential (active and passive) and
the IC50 after 24 h treatment was 0.0509 mg/mL for Taxotere, most importantly oral deliverability [65].
0.00658 mg/mL for the non-targeted LPNPs formulation and Recently, the stability of the PTX loaded lipid nanocapsules was
0.00323 mg/mL for the targeted LPNPs formulation. Thus, targeted established in the gastrointestinal tract. The stability of these systems
LPNPs formulation was 50.91% more effective than the non-targeted was attributed to the presence of the surface PEG coating, which specif-
LPNPs formulation and 93.65% more effective than Taxotere. ically protected the aggregation of internal lipid constituents due to
Joshi et al. developed and evaluated hybrid lipopolymeric system acidic pH in the stomach and against pancreatin activity in intestine.
comprising carboxymethyl chitosan (CMC), covalently tethered to However, weaker protection was exhibited in the presence of bile
phosphatidylethanolamine units on the surface of lipid nanovesicles, salts and hence pre-prandial administration of lipid nanocapsules for
for oral delivery of PTX. The bioploymer was intended to act as a blan- better stability prole along the gastrointestinal tract could be sought
ket, thereby shielding the drug from harsh gastrointestinal conditions, [66]. With these promising results, the uptake of the lipid nanoparticles
whereas the lipid nanovesicle ensured high encapsulation efciency of across the in vitro Caco-2 cell monolayer model was evaluated. About
PTX and its passive targeting to tumour. CMC-conjugated nanovesicles, 3.5-fold increase in the apparent permeability of PTX upon its loading
upon oral administration in rats, improved the plasma concentration in lipid nanocapsules was observed as compared to free drug; vesicle
prole of PTX, with 1.5-fold increase in its bioavailability and 5.5 folds' mediated transcytosis being the proposed mechanism for improved
increase in elimination half-life in comparison with Taxol. Also, CMC permeability [32]. These results were in correlation with pharmacoki-
in addition to providing a gastric resistant coating also imparted stealth netic studies conducted by another group. PTX-loaded lipid
character to the nanovesicles, thereby reducing their reticuloendotheli- nanocapsules demonstrated a 3-fold increase in the oral bioavailability
al system (RES)-mediated uptake by liver and spleen and bypassing the of PTX in comparison with Taxol orally administered. This effect on
need for PEGylation. In vivo efcacy in subcutaneous model of B16F10 the oral PTX bioavailability was directly related to the presence of the
showed signicantly improved tumour growth inhibition and survival surfactant Solutol HS15, which inhibited the P-gp efux system. Be-
with CMC-tethered nanovesicles as compared with unmodied sides, lymphatic transport of PTX and the nanocapsule structure might
nanovesicles, both administered orally. LN-C-PTX exhibited therapeutic also be benecial for the oral permeability of PTX [67].
efcacy comparable to Taxol and Abraxane and also showed reduced
toxicity and improved survival. 3. Polymer nanocarrier based approach
Jain et al. prepared AmB loaded polymer lipid hybrid nanoparticles
(AmB-PLNs) comprised of lecithin (anionic lipid) and gelatin (Type A, Polymer based nanocarriers have shown their potential to in-
cationic below its isoelectric point 7.09.0) [63]. The developed formu- crease the oral and peroral delivery of various drugs [6870]. Sub-
lation exhibited a sustained drug release prole with a release pattern stantial efforts have been made to improve the bioavailability vis-
best tted to Higuchi kinetics. Experiments on Caco-2 cell lines revealed -vis therapeutic efcacy and safety prole. A variety of biopharma-
a 5.89-fold increase in the intestinal permeability of AmB-PLNs whereas ceutical parameters have been reviewed to manipulate the in vivo
in vivo pharmacokinetic studies exhibited a 4.69-fold increase in the oral fate upon administration [71,72]. Briey, particle size, shape and sur-
bioavailability upon incorporation of AmB into PLNs as compared to that face properties of the nanoparticles play a crucial role in the uptake
of free drug. The developed formulation showed signicantly lesser across the gastrointestinal membrane and were found to signicant-
heamolytic toxicity as compared to the free drug, Fungizone (micellar ly affect the absorption prole. The nanocarriers with particle size of
solution of AmB) and Fungisome (liposomal formulation of AmB). Fur- 50300 nm, positive zeta potential and hydrophobic surface were
thermore, blood urea nitrogen (BUN) and plasma creatinine levels, in- found to have preferential uptake from gastrointestinal tract as com-
dicative of nephrotoxicity, were also found to be signicantly lesser pared to their counterparts. However, the retention of these proper-
for developed PLN formulation as compared to free drug and Fungizone ties in the gastrointestinal lumen is equally important and efforts
while comparable to that of Fungisome. The histopathology of the kid- should be made in this direction to maximize the delivery efciency
ney tissues further conrmed the absence of any changes in the mor- of the carrier system these properties in the gastrointestinal lumen is
phology of the renal tubules. equally important.
Sambaraj et al. [64] formulated and evaluated a novel silica-lipid hy-
brid (SLH) microparticulate system for improved oral delivery of FUR. 3.1. Mechanism of improved bioavailability of BCS class IV drugs by poly-
Silica-lipid hybrid microparticles included the drug solubilizing effect mer nanocarrier based approach
of dispersed lipids and stabilizing effect of hydrophilic silica particles
to increase drug solubilization, which lead to enhanced oral bioavail- The principal advantages of polymeric nanocarriers include their
ability. In vitro dissolution studies conducted under simulated gastric increased solubilization potential, superior encapsulation, altered
medium revealed 2 to 4-fold increase in dissolution efciencies for absorption pathways, prevention of metabolic degradation within
SLH microparticles compared to that of pure drug and marketed formu- gastrointestinal tract, chemical versatility of materials eligible for
lation Lasix. SLH also showed enhanced permeability when compared nanomedicines, exibility in surface functionalization, drug and dis-
to other formulations and marketed drug. The results indicated that that ease specic tailor made design capability, targeting potential and
SLH microparticles were better than pure drug and marketed drug ability to incorporate wide variety of drug substances. Nanocarriers
formulation. by the virtue of their ability are able to bypass the different hurdles
that are responsible for poor oral absorption of majority of BCS
class IV drugs. Various identied absorption mechanisms through
2.2.7. Lipid nanocapsules which nanocarriers increase the oral bioavailability of drug mole-
Lipid nanocapsules are the novel colloidal nanocarriers having typi- cules include increased absorption from enterocytes (due to in-
cal core shell arrangement of the constituents with liquid reservoir creased solubilization and dissolution), mucoadhesion (interaction
(usually oils) as core coated by protective membrane. The protective between the positively charged nanocarrier with negatively charged
membrane generally comprises of polymer chains present in the surfac- mucin), tight junction modulation (capability of nanocarriers to in-
tant which is aligned as exterior coat during the preparation of lipid teract with the tight junction proteins), receptor mediated endocy-
nanocapsules. The lipid nanocapsules are principally known for their tosis and transcytosis (clathrin- and calveolae-dependent and
biocompatibility, biodegradability, superior encapsulation of drug mol- -independent endocytosis), phagocytosis via specialized microfold
ecules, very high drug payloads, specic absorption mechanism across cells (M cells) of the Peyer's patches and other mucosa associated
the gastrointestinal tract, sustained release, membrane efux lymphoid tissues (MALT).
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 79

3.2. Potential applicability of polymer based nanocarriers for improving the pose potential delivery challenges. The drug molecules can be either
bioavailability of BCS class IV drugs physically entrapped or chemically conjugated to the dendritic struc-
tures during or after synthesis of macromolecular systems. Further-
3.2.1. Polymeric micelles more, encapsulation of drug molecules within the dendritic structures
Polymeric micelles are the systems containing hydrophobic cores selectively eliminates solubility and permeability related issues.
surrounded by hydrophilic corona that is exposed to aqueous environ- Jain et al. developed muramyl dipeptide (MDP) conjugated
ment. The hydrophobic cores act as a reservoir for lipophilic drug mole- multimeric poly(propyleneimine) (PPI) dendrimers for targeted deliv-
cules and corona acts as the steric stabilizer of the overall system ery of AmB to macrophages [98]. Therapeutic activity and toxicity of
thereby assuring the integrity of the system in aqueous environment dendrimeric formulation of AmB (MdPPIA) were compared with
holding the adequate amount of guest drug molecules [73]. Polymeric marketed formulations of AmB. Highly signicant (P b 0.01) reduction
micelles have recently gained wide acceptance as the carrier systems in toxicity was observed in hemolytic toxicity and cytotoxicity studies
considering their various advantages such as superior stability com- in erythrocytes and J774A.1 macrophage cells, respectively. Formulation
pared to surfactant micelles, enhanced solubilizing power, longer circu- MdPPIA showed appreciable macrophage targeting potential in cell up-
lating time due to outer hydrophilic shell, small size and targeting take studies. The developed formulation MdPPIA showed higher or
capability [74]. equivalent antiparasitic activity against parasite infected macrophage
Shaor et al. designed a formulation of 1, 2-distearoyl-sn-glycero- cell lines and in vivo infection in Balb/c mice.
3-phosphoethanolamine-N-[methoxy(polyethylene glycol)- 2000] Teow et al. investigated the ability of a third-generation (G3)
(PE-PEG) based micelles loaded with antimicrobial agent, AmB, and polyamidoamine (PAMAM) dendrimer-based carrier to enhance the
surface-modied with angiopep-2. The targeted micelles exhibited permeability of PTX and to overcome cellular barriers [99]. G3
a signicantly enhanced ability to carry AmB into the brain. In addi- dendrimers were surface modied with lauryl chains (L) and conjugat-
tion, the micellar AmB formulations demonstrated decreased cyto- ed with PTX via a glutaric anhydride (glu) linker, followed by labeling
toxicity and hemolysis in vitro compared with Fungizone [75]. PTX, with FITC. Biological evaluation of the dendrimer and conjugates was
with an aqueous solubility of approximately 0.3 g/mL, was loaded conducted using the Caco-2 cell line and primary cultured porcine
into 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N- brain endothelial cells (PBECs). Cytotoxicity studies showed that the
methoxy(PEG)/TPGS (PEG-DSPE/TPGS) micelles. The aqueous solu- conjugation of L and PTX on G3 dendrimer signicantly (p b 0.05) in-
bility of PTX was enhanced by up to 5000 times to achieve an aque- creased the cytotoxicity against both cell types. Permeability studies of
ous solubility of approximately 5 mg/mL [76]. The most impressive dendrimerdrug conjugates demonstrated an increase in the apparent
enhancement in PTX solubility was achieved by the work of Kim et permeability coefcient (Papp) in both apical to basolateral A B and
al., in which it was reported that the aqueous solubility of PTX was basolateral to apical B A direction across both cell monolayers com-
as high as 38.9 mg/mL through encapsulation in micelles. In this pared to unmodied G3 and free drug. L6- G3-glu-PTX conjugate had
study, nicotinamide derivatives, i.e. N, N-diethylnicotinamide and approximately 12-fold greater permeability across both cell monolayers
N-picolylnicotinamide (DENA) were shown to be powerful than that of PTX alone.
hydrotropes for PTX. Copolymers with a segment containing such Devarakonda et al. investigated the efciency of lower generation
nicotinamide derivatives could be used to produce polymeric PAMAM dendrimers to improve the solubility and release kinetics of
micelles with hydrotropic properties toward PTX. In addition, FUR [100]. The increase in the solubility of FUR in the presence of
micelles composed of PEG-b-poly(vinylbenzyloxy)- N, N- dendrimers was primarily due to the electrostatic interactions between
diethylnicotinamide (PEG-b-PVBODENA) could achieve a remark- the positively charged tertiary amines of the dendrimers and negatively
ably high drug loading (37.4% w/w) for micelle-based delivery charged carboxylate anion of FUR, resulting in a favourable solubiliza-
systems. The drug loading increased proportionally to the length of tion effect. The results showed that the PAMAM dendrimers could be
the hydrotropic DENA segment. As a comparison, PEG-b- PLA mi- exploited to improve the solubility and dissolution of FUR, however
celles could only load up to 27.6% (w/w) of PTX under similar condi- the enhancement depended on the choice of dendrimer, generation
tions [77,78]. size, and surface functional group of the dendrimer.

3.2.2. Polymeric nanoparticles 4. Pharmaceutical crystal engineering


Polymeric nanoparticles are nano-colloidal cargos, preferably in the
size range of 101000 nm, made up of wide variety of polymers. They Crystal engineering is a new and emerging method of controlled
have been widely studied and evaluated for the oral and per-oral deliv- crystallization that can be described as the exploitation of noncovalent
ery of many drugs and chemotherapeutic agents. The principal advan- interactions between molecular or ionic components for the rational de-
tage of this system includes their robust structural characteristics sign of solid-state structures. Crystal engineering technologies can be
imparting very high stability in the gastrointestinal tract. Furthermore, applied to pharmaceutical substances like BCS class IV drugs to improve
the hydrophobicity and hydrophilicity within the polymeric system drug solubility and permeability through controlled crystallization pro-
can be manipulated to accommodate wide variety of drug molecules cesses such as by forming co-crystals, nanocrystals, lipid nanocrystals,
[79]. The properties of biocompatibility and biodegradation further en- metastable polymorphs, high energy amorphous forms and ultrane
hance their delivery potential. A large number of polymers including the particles.
co-polymers have been employed for the preparation of polymeric
nanoparticles (nanocapsules and matrix based nanoparticles). Table 2 4.1. Nanocrystal technology
reveals various types of polymeric nanoparticles that have been imple-
mented to improve the oral and per oral delivery of various BCS class IV Nanocrystals, is currently offering promising potential to address the
drugs. low solubility and poor bioavailability problems of many drug candi-
dates [101]. A drug nanocrystal is a nano-sized crystalline particle con-
3.2.3. Dendrimers taining 100% drug with nominal utilization of excipients like surface
Dendrimers are the hyper branched and uniformly distributed mac- stabilizing agents. The pharmaceutical benets of nanocrystals include
romolecules that possess denite molecular weight, shape, size and spe- improvement in formulation performance, such as enhanced dissolu-
cic chemical and physical properties including hostguest entrapment tion velocity and saturation solubility due to increased surface area/vol-
properties [97]. The tailor made design makes these sophisticated sys- ume-ratio [102]. Nanocrystal formulations exhibit a high drug load;
tems eligible for incorporation of wide variety of drugs that otherwise reproducibility of oral absorption, improved dose-bioavailability and
80 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

Table 2
Polymeric nanoparticles implemented to improve the oral and per oral delivery of BCS Class IV drugs.

Polymeric system
(Functionalization) Drug Outcome References

PAA-cysteine (GSH) PTX About 7-fold increase in apparent permeability and about 5-fold increase in oral bioavailability as compared to free paclitaxel [80]
Polyanhydrides PTX Relative Bioavailability up to 80% [81]
(Cyclodextrins)
Polyanhydrides (PEG PTX Relative bioavailability up to 70% [82]
2000)
PLA (Vitamin E TPGS) DTX Bioavailability increased by 22-fold [83]
PLA (TPGS) PTX 1.8-Fold increase in the cellular uptake by HT29 cell lines as compared to plain PLGA nanoparticles and 40% reduction in IC50 [84]
values as compared to taxol
PLGA (DMAB-TPGS) DTX About 16-fold decrease in the IC50 value for in vitro cytotoxicity against MCF-7 cell lines as compared to marketed formulation [85]
at 72 h
PLGA (Montmorillonite) PTX Signicant improvement in the entrapment efciency by 57177% for Caco-2 cells and 1155% for HT-29 cells was observed [86]
PLGA (WGA) PTX About 2-fold increase in the cellular uptake by Caco-2 and HT-29 cell lines as compared to plain nanoparticles [87]
PLGA (WGA) PTX Signicantly higher cytotoxicity against A549 and H1299 cell lines [88]
PLGA PTX 7-Fold higher toxicity of formulation in A2780 cell lines and signicant activity in 2780 CE resistant cell lines [89]
PLGA (Mannose) AmB Enhanced uptake, potential anti-leishmanial activity and higher disposition in macrophage-rich organs, suggesting improved [90]
macrophage targeting
Pullulan acetate LPV Relative bioavailability of LPV from nanoparticles was 2 folds higher than the free drug, higher distribution of nanoparticles to [91]
lymphoid organs
PCL LPV Increase in oral bioavailability of LPV by 4-folds [92]
PLGA (TPGS) AmB Lower haemolysis, nephrotoxicity and relative oral bioavailability was found to be 800% as compared to Fungizone. [93]
PLGA AmB Superior in vitro antifungal activity (two times more efcacious than AmBisome) [94]
PLGA CyA Signicantly higher intestinal uptake as compared to Sandimmune Neoral and cyclosporine suspension, relative [95]
bioavailability of nanoparticulate formulation was found to be 119.2%
PLGA (Folic acid) SQV Enhanced cytotoxicity and cellular uptake by the cancerous cells via folate receptor-mediated endocytosis (RME) mechanism [96]

PLGA: Polylactide-co-glycolic acid; PCL: poly--caprolactone; PLA: Polylactic acid; PEG: Polyethylene glycol; WGA: Wheat germ agglutinin; PAA: polyacrylic acid; GSH: glutathione.

versatile administration routes along with proportionate and increased nanocrystals are recognized as foreign bodies and engulfed by the de-
patient compliance. Also reduction of number of oral units to be taken, fence (phagocytic) cells such as macrophages. In phagocytic cells
low incidence of side effects due to the excipients and an overall im- nanocrystals dissolve slowly in phagolysosomes. Consequently, the hy-
provement of efciency and safety can also be achieved. drophobic drug might pass through the phagolysosomal membrane and
enter into the cytoplasm, and then diffuse out of the cell down its con-
4.1.1. Mechanism of improved bioavailability by nanocrystal technology centration gradient [108]. Fig. 4(C) depicts the fate of nanocrystal after
Nanocrystals and their uptake are not fully understood although intravenous administration. Nanocrystals of size b100 nm behave like
several studies have reported various mechanisms as well as pathways. molecular solution whereas those N 500 nm are accumulated in the
Paracellular pathway is a route for potential uptake of nanocrystals. liver. This characteristic of nanocrystals can be successfully used in
However, owing to the strict diametric constriction (b 10 ) of the targeting different diseases/disorders [109].
pores present in between juxtaposed cells, exponentially larger
nanocrystals (~100 nm onwards) nd it difcult to pass through. Fig. 4.1.2. Potential applicability of nanocrystal technology to BCS class IV drugs
4(A) illustrates paracellular pathway of enterocytes which hinders Oral administration of AmB as a nanosuspension produced a sub-
transport of nanocrystals. However, some researchers claim the open- stantial improvement in its oral absorption in comparison to orally ad-
ing of these junctions by the application of some specic chemicals (sur- ministered conventional commercial formulations such as Fungizone
factants) such as sodium deoxycholate which might facilitate the entry and AmBisome. Apart from improving oral absorption, nanosus-
of nanocrystals, but there is no adequate experimental evidence on this pensions offered improved dose proportionality, reduced fed/fasted
[103]. In case of cancer the paracellular uptake at site of action may take state variability and reduced inter-subject variability [111]. The devel-
place owing to the well-known enhanced permeability and retention opment of PTX nanocrystals for oral delivery has recently been often
(EPR) effect (high fenestra frequency) [104]. Some nanocrystals are proposed as a promising approach to overcome the drawbacks of classic
taken up directly by cells via multiple endocytotic pathways including products on the market. For instance, several studies have considered
clathrin-mediated, non-clathrin mediated (caveolae), macro-pinocyto- the possibility of modifying the nanocrystal surface in order to enhance
sis, as well as phagocytosis [105]. Fig. 4(B) demonstrates the above spe- their properties. To this end, Patel et al. [112] focused their investigation
cialized mechanisms of nanocrystal transport via enterocytes. After on developing a PTX nanocrystal formulation in order to overcome the
endocytosis nanocrystals possibly become available for lymphatic drug's poor solubility and permeability. In particular, positively charged
transport instead of venous transport. The reasons might be the low nanocrystals were prepared by sonoprecipitation with a high-pressure
pore size (roughly 3 nm) of the vascular epithelium. Reports on the up- homogenization method to investigate a combination of arginine
take of nanocrystals via M-cells are also found in literature. These spe- based cationic surfactant and hydroxypropylmethylcellulose. Both the
cialized cells are believed to transport nanocrystals through an active dissolution and permeability of PTX were signicantly improved by
transepithelial vesicular transport system from the lumen directly to nanocrystal formulation, and the in vitro and in vivo results clearly sug-
lymphoid cells and tissues; though the overall efciency of this process gested it to be a promising approach in the anticancer eld.
remains questionable as M cell populates b 1% of the intestinal epithelia Also to augment passive targeting, ligands selective for receptors
[106]. over expressed on cancer cells are appended to nanocrystals. This
Following intravenous administration, nanocrystals may remain in- opens up the avenue for active receptor based targeting by tailoring
tact or enter into a molecular dispersion due to high intrinsic solubility, nanocrystals [113]. Liu et al. described the concept of proof by activating
dissolution rate, and rapid dilution accessing the site of action for ther- PTX nanocrystals through application of folic acid conjugated stabilizer.
apeutic effect [107]. Alternatively, intact nanocrystals keep moving in In comparison to non-ligated PTX nanocrystals, ligated nanocrystals
the systemic circulation as colloidal particles. During their ight, showed enhanced in vitro cytotoxic activity due to preferential uptake
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 81

Fig. 4. (A) Graphical representation of intracellular tight junction and failure of nanocrystals to pass through paracellular pathway, (1) enterocytes, (2) intracellular tight junction (b10 ),
and (3) failure of nanocrystals to pass through the paracellular pathway, (4) uptake of nanocrystals by enterocytes after treatment with surfactants, and (5) M-cell mediated uptake of
nanocrystals. (B) Diagrammatic representation of clathrin mediated, and non-clathrin (caveolae) mediated endocytosis of nanocrystals after oral administration, (1) nanocrystals on
the enterocyte surface, (2) magnied view of nanocrystals over the cell membrane, (3) clathrin-mediated phagosome and endocytosis of nanocrystals, (4) release of nanocrystals to
the other side of cell via clathrin-mediated transport, (5) nonclathrin (caveolae) mediated endocytosis of nanocrystals, and (6) release of nanocrystals to the other side of cell via non-
clathrin (caveolae) mediated transport. (C) Proposed in vivo fate of nanocrystals after intravenous administration (1) injection of nanocrystals to blood vessels, (2) nanocrystals in the
blood stream in molecular solution and intact nanocrystal form, (3) phagocytic cells, (4) formation of molecular solutions of nanocrystals in blood, (5) phagocytosis of intact
nanocrystals and further solubilization in cytosol of phagocytes, and (6) drug in molecular solution form comes out due to concentration gradients of phagocytes and outer
environment. Adopted and modied from [110].

via over expressed folic acid receptor on KB cells [114]. Puligujja et al. vivo delivery of BCS Class IV drugs. It is a unique approach to solve
produced long-acting folic acid-receptor targeted RTV-boosted bioavailability related issues. Industrially feasible manufacturing
atazanavir nanoformulations to improve the antiretroviral therapy techniques can serve to bring nanocrystals from researcher's bench
[115]. Folic acid was covalently linked to two types of poloxamers, to patient's bed at an accelerated rate. Nanocrystals are versatile
that is, poloxamers 407 and 188. Subsequently, the conjugate was pharmaceuticals which can be delivered through almost all routes
used to nanoencapsulate antiretroviral drug nanocrystals. These coated of administration such as oral, parenteral, dermal, and ocular. They
nanoparticles targeted the folic acid receptor favouring drug tissue de- are adept to convenient sterilization techniques, a key driver for de-
pots and permitting an enhanced accumulation to the reticuloendothe- velopment of parenteral formulations. Incorporation of appropriate
lial system with an improved pharmacodynamics activity in mice. stabilizers followed by drying (freeze/spray) can ensure long term
Table 3 depicts the various BCS class IV drugs for which the nanocrystal stability of nanocrystals. Nanocrystals decorated with functionalized
technology led to increased bioavailability and therapeutic potential. ligands have generated a new ray of hope by being able to target var-
Taking into account potential advantages and reduced side ef- ious organs with higher afnity. Furthermore, nanocrystals might be
fects, nanocrystals can be considered as suitable candidates for in- an opportunity to produce generic versions of presently available
82 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

Table 3
Nanocrystals implemented to improve the bioavailability of BCS Class IV drugs.

Drug (route) Therapeutic use Benet Reference

PTX (i.v.) Cancer Signicant anti-tumour efcacy of nanocrystals over Taxol [116]
PTX & Cancer Enhanced antitumour activity due to targeting folate receptors [114]
camptothecin
(oral & i.v.)
DTX (i.v.) Cancer DTX nanocrystals proved a better opportunity to inhibit tumour growth and reduce toxicity in comparison to [117]
marketed formulation.
AmB Organ targeting (Brain) Nanocrystals coated with polysorbate 80 and sodium cholate showed enhanced brain uptake than AmB [118]
solution
Aprepitant Chemotherapy induced The fed-fasted ratio was reduced and the bioavailability was improved in the beagle dogs at a dose of 2 mg/kg [119]
nausea and vomiting
RTV HIV The nanosuspension showed a signicantly increased solubility when compared to coarse drug powder (3.5 [120]
fold), 90% drug dissolved in FeSSIF compared to the commercial product which showed ~50% in the same
medium
RTV HIV Improved inhibition of CYP3A4, enhanced permeability across Caco-2 cells lower cytotoxicity across hepatic, [121]
intestinal, and immune cell types by nanodispersions compared to an aqueous solution of RTV.
LPV HIV The surface-stabilized LPV-NPs (without RTV) demonstrated a 3.11-fold enhancement in bioavailability in [122]
comparison to free LPV with RTV (conventional formulation).

costly drugs. In conclusion, engineered nanocrystal technology has liquid on its surfaces is called the carrier which usually has the highest
huge potential to deliver pre-existing or newly developed notorious contribution in the formulation. The excipient with very high surface
class IV drugs in a more acceptable and effective dosage form with area which usually covers the carrier surfaces containing liquid to im-
high commercial applicability. prove owability of liquisolid powders is known as coating materials.
The outline of liquisolid preparation is presented in Fig. 5.
4.2. Co-crystal technology
5.1. Mechanism of improved bioavailability of BCS class IV drugs by
Recently, pharmaceutical co-crystals have gained interesting atten- liquisolid technology
tion to act as drug candidates in solid-state drug development. The mo-
lecular components of co-crystal complexes are the target molecule In the literature, several mechanisms of enhanced drug release have
(traditionally the drug molecule) and the co-crystal former(s) (also been postulated for liquisolid systems. The three main suggested mech-
known as the co-crystallizing agent or co-former). Co-crystals are anisms include an increased surface area of drug available for release,
formed by intermolecular interactions, or synthons, that form between increased solubility of the drug and improved wettability of the drug
the drug and a potential co-crystal former, which in turn leads to the particles.
creation of supramolecular assemblies. A pharmaceutical co-crystal
does not only provide new opportunities to modify the physicochemical 5.1.1. Increased drug surface area
properties, dissolution rate, and drug bioavailability of an API, but also Even if the drug within the liquisolid system is completely dissolved
creates intellectual property for a new patent of an API for extending in the liquid vehicle, it is still located in the powder substrate in a solu-
their life cycle. Thus, the investigations of pharmaceutical co-crystals bilized, molecularly dispersed state. Therefore, the surface area of drug
have been rapidly expanded by growing the number of research publi- available for release is much greater than that of drug particles within
cations and patent applications [123]. directly compressed tablets. Accordingly, with increasing drug content
exceeding the solubility limit and thus increasing fraction of undis-
solved drug in the liquid vehicle the release rate decreases. With various
4.2.1. Potential applicability of co-crystal technology to BCS class IV drugs
drugs, it could be shown that the release rates are directly proportional
Sanphui et al. addressed the poor solubility of the diuretic drug HCT
to the fraction of the molecularly dispersed drug (FM) in the liquid for-
by preparing co-crystals [20]. The co-crystal of HCT with p-
mulation [125127]. FM is dened by Spireas et al. as the ratio between
aminobenzoic acid showed a six-fold increase in solubility compared
the solubility (Sd) in the liquid vehicle and the actual drug concentra-
with pure HCT, and stability up to 24 h in an aqueous medium. The
tion (Cd) in this vehicle carried by each system [126]. Therefore:
co-crystals of HCT with nicotinamide, resorcinol and pyrogallol showed
a 1.5 to 2- fold increase in solubility. Goud et al. reported a co-crystal of FM Sd=Cd
FUR, a low soluble and low permeable antihypertensive drug with caf-
feine using liquid assisted grinding [124]. The co-crystals with improved where FM = 1 if Sd = Cd.
physicochemical properties exhibited higher solubility, more stability
and increased dissolution rates than the pure compound. The solubility 5.1.2. Increased drug solubility
and intrinsic dissolution rate for the co-crystal was found to be 6 and 2 In addition to the rst mechanism of drug release enhancement, it is
times more respectively, than FUR. expected that Cs, the solubility of the drug, might be increased with
liquisolid systems. In fact, the relatively small amount of liquid vehicle
5. Liquisolid technology in a liquisolid compact is not sufcient to increase the overall solubility
of the drug in the aqueous dissolution medium. However, at the solid-
Liquisolid systems are considered as free-owing and compressible liquid interface between an individual liquisolid primary particle and
or compactable powders containing a non-volatile liquid vehicle and the release medium, it is possible that in this microenvironment the
solid drug particles. The solid drug particles can be dissolved or partly amount of liquid vehicle diffusing out of a single liquisolid particle to-
dissolved by the liquid vehicle (co-solvent). This liquid system may be gether with the drug molecules might be sufcient to increase the solu-
converted into a dry looking, free-owing and compressible powder bility of the drug if the liquid vehicle acts as a co-solvent. The overall
by mixing with suitable excipients termed the carrier and coating mate- increase in the solubility of drugs caused by liquisolid systems was con-
rials. In liquisolid formulations, the excipient with a capability to adsorb rmed by Yadav et al. [128130].
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 83

Fig. 5. Liquisolid technology for improving the bioavailability of BCS Class IV drugs, (A) The components of a liquisolid system, (B) method for the formulation and development of
liquisolid system, (C) Potential advantages and outcomes of liquisolid system.

5.1.3. Improved wetting properties addition, the selected optimized formula released 78.36% of its content
Due to the fact that the liquid vehicle can either act as surface active during the rst 10 min which is 39% higher than that of the directly
agent or has a low surface tension, wetting of the liquisolid primary par- compressed tablets. Further, the bioavailability study indicated that
ticles is improved. Wettability of these systems has been demonstrated the prepared optimized liquisolid formula did not differ signicantly
by measurement of contact angles [131] and water rising times [128, from the marketed FAM tablets concerning Cmax, tmax, and AUC (08) at
130]. Liquisolid compacts show lower contact angles compared to con- P b 0.05. Akinlade et al. enhanced the in vitro dissolution properties of
ventional tablets due to the presence of a surface active agent, for exam- FUR, by utilising liquisolid technique [133]. Liquisolid tablets containing
ple, polysorbate 80 [127,131]. Synperonic PE/L 81 as a new liquid vehicle exhibited greater dissolu-
tion due to the physical properties of this liquid vehicle which led to in-
creased wetting properties and solubility of the drug; demonstrating
5.2. Potential applicability of liquisolid technology to BCS IV class drugs higher drug release than those of conventionally made tablets by direct
compression.
The technique of liquisolid compacts has been successfully Khaled et al. evaluated the absorption characteristics of experimen-
employed to improve the in vitro release prole of many poorly water tally developed HCT liquisolid tablets using six male beagle dogs [136].
soluble drugs. Further, this technique has also been tried to improve Signicant differences were found between the two formulations i.e.,
the bioavailability of many BCS class IV drugs like FAM [132], FUR liquisolid tablets and commercial tablets with regard to AUCt, AUC,
[133,134], CyA [135] and HCT [136]. Fahmy et al. formulated FAM Cmax, and F parameters. Liquisolid tablets consistently showed higher
liquisolid systems to improve its dissolution and also investigated the values of the aforementioned parameters. The mean values of the abso-
in vitro and in vivo performance of the prepared liquisolid tablets lute bioavailability of HCT from the commercial and the liquisolid tab-
[132]. All the tested liquisolid tablet formulations showed higher drug lets were found to be 72.6 and 83.7%, respectively and signicantly
dissolution rates than the conventional, directly compressed tables. In different. This nding reects an increase in the bioavailability of the
84 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

drug by 15% when formulated as liquisolid tablets. The Cmax value toxicological evaluations and precedence of use in approved pharma-
achieved following the administration of the liquisolid tablets was ceutical products.
32% higher than that of the commercial tablets. The mean absorption
time for the liquisolid tablets was 1.58 h, which was 55 min shorter 6.1.2. Tocopheryl polyethylene glycol 1000 succinate (TPGS)
than the commercial tablets (2.5 h). Although this difference was not Another lipid-based excipient useful for preparing self-emulsifying
statistically signicant, this reected the rapid absorption of the solid dispersion formulations is FDA approved water soluble d--
liquisolid tablets. The parametric 90% condence intervals of the mean tocopheryl polyethylene glycol 1000 succinate (TPGS; Vitamin E TPGS,
values of AUCt, AUC, and Cmax were 107.5132.2, 102.8131.7, and Eastman Chemical, Tennessee, U.S.A.). This excipient, which is listed in
110.7153.7%, respectively. These intervals are, generally, higher than the United States Pharmacopoeia, is a water-soluble, surface-active,
the expected interval range for bioequivalency (80125%, using log and self-emulsifying derivative of vitamin E that is capable of solubiliz-
transformed data). This should be expected as liquisolid tablets showed ing many poorly water-soluble drugs. Apart from being a very good
greater extent of absorption than the commercial tablets. In conclusion, emulsier, it also has P-glycoprotein (P-gp) inhibiting property. It is
HCT liquisolid tablets showed signicantly greater extent of absorption being increasingly employed to overcome bioavailability issues associ-
than the commercial tablets. Zhao et al. tried to enhance the dissolution ated with poorly soluble compounds especially in case of anticancer
rate of CyA, a poorly water-soluble drug by developing a self-micro- drugs where it helps to overcome the MDR. Also, because of its relatively
emulsifying (SME) tablet formulation by using the liquisolid compact low melting point (~40 C), molten TPGS formulations are suitable for
technique [135]. Microcrystalline Cellulose (Avicel PH 101 and Avicel lling into both soft and hard gelatin capsules; however, hard gelatin
PH 102) and Magnesium Aluminometasilicate (Neusilin S1) were se- capsules require band-sealing in order to prevent leakage of the
lected as the carrier material and coating material respectively for pre- contents.
paring the liquisolid compact. The obtained powder mixture had good
owability (Hausner's Ratio = 1.243, Carr's Index = 19.565) and good 6.1.3. Pluronics
compactability (Hardness = 5.18 0.33kp, Tensile Strength = Another surface active lipid excipient class that has shown signi-
0.47 0.03Mpa). The dissolution results showed that the dissolution cant promise for application in self-emulsifying solid dispersion formu-
rate of SME tablets was much higher when compared to the conven- lations is the block copolymer consisting of hydrophilic poly(ethylene
tional tablets prepared by direct compression. oxide) (PEO) and hydrophobic poly(propylene oxide) (PPO), various
grades of which are commercially available as poloxamers (Pluronics)
6. Self-emulsifying solid dispersion formulations [139,140]. They possess both solvent and surfactant properties and suit-
able melting range which are well suited for capsule lling operations.
These formulations consist of a dispersion of the drug in an inert ex- They are well characterized with respect to quality, chemical compati-
cipient matrix, where the drug could exist in either the nely divided bility, and performance in promoting the absorption of hydrophobic
crystalline, solubilized or amorphous states or a mixture thereof. This drugs through solubilization or increased wetting of the drug particles.
can increase the dissolution rate of the drug in, and subsequent absorp- They also enhance cell membrane permeability and decrease intestinal
tion from, the GI tract relative to the stable crystalline drug substance. efux through inhibition of the P-gp transporter.
The introduction and use of newer thermo-softening, surface-active
and self-emulsifying excipients (Examples in Section 4.1) have 6.2. Mechanism of improved bioavailability of BCS class IV drugs by self-
surmounted the various issues that impeded the commercial develop- emulsifying solid dispersion formulations
ment of solid dispersions. These excipients have the potential to further
increase the absorption of poorly water-soluble drugs relative to previ- The lipid based commercially marketed products of CyA, RTV and
ously used PEG solid dispersions and may also be lled directly into hard SQV are encapsulated liquids. The primary limitation of these lipid-
gelatin capsules in the molten state, thus obviating the former require- based dosage forms is the requirement that the drug possess sufcient
ment for milling and blending prior to lling. In addition, more recently solubility in the formulation to allow delivery of a single dose in a stan-
introduced hot-melt extrusion technology has proven to be of value in dard oral capsule dosage form. In instances where insufcient solubility
resolving some of the remaining manufacturing issues associated with in the formulation precludes the development of a fully solubilized
solid dispersions. lipid-based formulation, preparation of a solid dispersion of the drug
in a semi-solid, lipid based formulation could provide a viable path for-
6.1. Examples of surface active excipients used in the fabrication of self- ward. However, the performance of non-self-emulsifying solid disper-
emulsifying solid dispersions sions (e.g., those prepared using PEGs) is often limited by the
relatively rapid dissolution rate of the water-soluble excipient matrix
6.1.1. Gelucires as compared to that of the dispersed drug substance. This results in
One excipient that has stimulated interest in self-emulsifying solid the formation of a highly concentrated solution of drug which precipi-
dispersion formulations is Gelucire 44/14 (Gattefoss Corp., St. Priest, tates on the surface of the dissolving excipient plug, forming a poorly
France). This self-emulsifying excipient exists as a waxy semi-solid at soluble coating that prevents further dissolution of dispersed drug
ambient room temperature and is a mixture of glyceryl and PEG 1500 contained within the excipient matrix. For this reason, solid dispersions
esters of long-chain fatty acids. The sufxes, 44 and 14, in the excipient must be pulverized and sifted to increase their surface area in order to
trade name refer to its melting point and hydrophiliclipophilic balance facilitate drug release. This is a difcult task as solid dispersions pre-
(HLB), respectively. Gelucire 44/14 has also been extensively used in pared with such common excipients as PEGs are usually soft and tacky
combination with other surfactants and solubilizing agents to enhance and may not be readily milled. Moreover, the powders thus produced
the performance of solid dispersions [137,138]. It has the ability to often have poor ow and mixing properties and are difcult to ll into
self-emulsify on contact with aqueous media forming a ne dispersion capsules or compress into tablets.
i.e. micro emulsion. Surfactive power improves the solubility and wetta- In comparison, solid dispersion formulations prepared from surface-
bility of the active pharmaceutical ingredients in vitro and in vivo. The active lipid or lipid like excipients prevent the formation of a poorly
increased bioavailability of the drugs could also be associated with the water-soluble drug surface layer on the excipient plug during dissolu-
inhibition of the enterocyte efux transporter (P-gp inhibition). It ex- tion. While a portion of the released drug may still precipitate in the dis-
hibits good thermoplasticity for use as a binder in melt granulation solution medium once its dissolved concentration exceeds the
and melt extrusion techniques, associated with rapid formation of sta- saturation solubility, it would typically be present in a nely divided
ble crystalline phase. The safety of use is supported by extensive state due to the surface active properties of the excipient. The associated
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 85

high surface area of the nely divided drug substance would facilitate its were used to prepare pellets. The dissolution study depicted that, the
rapid dissolution in the GI uid. This is shown schematically in Fig. 6. for presence of the drug in solid dispersion enhanced its dissolution in com-
solid dispersions lled into hard gelatin capsules. Another major advan- parison with the drug itself. Also, the drug release from the
tage of solid dispersion formulations prepared from lipid or surface ac- manufactured pellets was found to be improved in the case of solid dis-
tive excipients is realized in the relative ease with which they are persions (drug: carrier 1:3). A complete drug release occurred after
manufactured. Solid dispersions prepared from these excipients may 30 min from pellets containing solid dispersions, while only about 30%
be directly lled into hard gelatin capsules in the molten state, which of the loaded FAM was released from pellets containing untreated
subsequently solidify upon cooling to ambient room temperature, drug after 2 h.
thus eliminating the need for grinding, sifting, and mixing. The melting
temperature of the molten excipient, however, must not exceed ~70 C,
7. P-glycoprotein (P-gp) inhibition strategies
which is the maximum acceptable ll temperature for hard gelatin cap-
sules [141].
The intestinal efux pump, P-glycoprotein (P-gp), located in the api-
cal membranes of intestinal absorptive cells, can reduce the bioavail-
6.3. Potential applicability of self-emulsifying solid dispersion formulations
ability of a wide range of drugs which are substrates for this
to BCS class IV drugs
membrane transporter. The BCS class IV drugs which are substrates of
P-gp efux pumps mainly belong to the class of HIV protease inhibitors
Investigators have studied the role of TPGS in enhancing the absorp-
(IDV, SQV, LPV, NFV) and taxanes (PTX, DTX). Various formulation strat-
tion of drugs, which are both poorly soluble and poorly permeable
egies to effectively inhibit P-gp mediated efux of BCS class IV drugs
[142]. Sokol et al. [143] reported enhanced absorption of CyA from vita-
have been tried and evaluated as shown in Fig. 7. These strategies inde-
min E TPGS formulations administered to pediatric patients, as com-
pendently and in combination, are: (a) co-administration of another P-
pared to formulations without vitamin E TPGS. Formulations including
gp substrate/specic inhibitor, (b) incorporation of a nonspecic lipid
TPGS allowed a 40% to 72% reduction in the CyA dosage required to
and/or polymer excipient in the formulation which blocks the activity
maintain therapeutic plasma drug concentrations. Similarly, Chang et
of P-gp efux pump and (c) novel peptide prodrug approach. The rst
al. [144] reported a signicant increase in CyA bioavailability in healthy
approach, although very effective in inhibiting P-gp, utilizes a second ac-
volunteers when administered with TPGS. In both of these instances,
tive compound in the formulation and thus imposes regulatory con-
the investigators suggested not only drug solubilization, but also inhibi-
straints and long development timelines on such combination
tion of the intestinal P-glycoprotein transporter as a contributing factor
products. Excipient inhibitors appear to have minimal nonspecic phar-
to the enhanced CyA bioavailability.
macological activity and thus potential side effects of specic active
Sinha et al. developed self-emulsifying solid dispersion of RTV by dif-
compound inhibitors can be avoided. Case studies have been presented
ferent methods (solvent evaporation [SE], melt method [MM]) and in-
where specic active compounds, surfactants, polymers, and formula-
vestigated them for in vitro and in vivo performance for enhancing
tions incorporating these molecules are shown to signicantly improve
dissolution and bioavailability, respectively [145]. Since the drug pos-
the intestinal absorption of poorly soluble and absorbed BCS class IV
sessed food-related absorption, the effect of biorelevant media (fasted
drugs as a result of P-gp inhibition and enhanced drug transport in
[FaSSIF] and fed [FeSSIF] state simulated intestinal uid) on dissolution
vitro. (See Fig. 8.)
behaviour was also studied. RTV and Gelucire formed a eutectic mixture
in the ratio of 1:4. In vitro dissolution studies showed that release of RTV
from solid dispersions was more than that of pure drug, which was due 7.1. P-gp inhibition by lipidic and polymeric excipients and formulations
to the nano size of dispersion. When the dissolution was performed in
FaSSIF and FeSSIF media, maximum dissolution was obtained with There are many reports in the literature on the modulation of the ac-
FeSSIF media, which conrmed food-related absorption of drugs. The tivity of P-gp using lipidic and polymeric excipients. The following case
apparent rate of absorption of RTV from SE (Cmax 20,221.37 ng/mL, studies are representative of this rapidly growing eld of
tmax 0.5 h) was higher than that of MM1 (Cmax 2462.2 ng/mL, tmax biopharmaceutics and its applicability to BCS class IV drugs. Enhance-
1 h) and pure drug (Cmax 1354.8 ng/ml, tmax 0.5 h). The results indicated ment of PTX solubility and permeability by TPGS in vitro/in situ and ef-
that the solid dispersion is a promising approach for the bioavailability fects on in vivo drug absorption has recently been reported by Varma
enhancement of RTV and can be used for the solid dosage form develop- et al. [22]. In situ studies using rat ileum tissue in Ussing chambers, the
ment for oral use in order to commercialize. Ibrahim et al. [146] pre- bi-directional transport of 14CPTX was also monitored in the presence
pared solid dispersion of FAM solid using two hydrophilic carriers, of increasing concentrations of TPGS. The apical-to-basolateral (A B)
namely Gelucire 50/13 and Pluronic F-127 and the solid dispersions permeability of PTX was found to increase in the presence of TPGS
while the basolateral-to apical (B A) was decreased with maximum
effect at 0.1 mg/mL of TPGS. Increasing TPGS concentrations above
0.1 mg/mL, resulted in a decrease in A B permeability with no change
in the B A permeability, which suggests the involvement of mono-
meric and not micellar TPGS in P-gp inhibition [22]. Wasan et al. [147]
conducted studies to ascertain that the incorporation of (AmB) into a
glyceride-rich excipient Peceol (glyceryl monooleate) signicantly in-
creased gastrointestinal absorption of AmB in white male Sprague-
Dawley rats. Based on preliminary studies, [147] it was proposed that
incorporation of AmB into mixed micelles composed of Peceol would
signicantly enhance gastro-intestinal (GI) tract absorption by decreas-
ing P-gp mediated drug efux. A signicant lower MDR-1 mRNA and P-
gp protein expression within Caco-2 cells was observed following 1-
and 7-day treatment with Peceol 0.1% to 1.0% (v/v) compared to non-
treated controls [148]. Taken together, these ndings suggested that
Peceol increased the gastrointestinal absorption of AmB by decreasing
MDR-1 mRNA and P-gp protein expression, resulting in lower PGP-me-
Fig. 6. Potential advantages of self-emulsifying solid dispersions. diated AmB efux [148].
86 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

Fig. 7. Various P-gp based approaches to improve the bioavailability of BCS Class IV drugs. (A) Potential mechanism for the inhibition of P-gp efux pump. (B) Strategies to inhibit/modulate
P-gp efux pump.

Zhang et al. prepared polymeric micelles based on monomethoxy monolayers showed that the Papp was signicantly increased when
poly(ethylene glycol)-b-poly (D, L-lactic acid) (mPEG-PLA) to improve CyA was formulated with the copolymer below its critical association
the oral bioavailability of CyA [149]. In vitro release test showed that concentration (CAC) and no signicant difference was found above its
the cumulative release percentage, about 85%, of CyA from polymeric CAC, implying that mPEG-PLA monomers affected the intestinal P-gp ef-
micelles within 24 h was comparable to that from Sandimmune Neoral, ux pumps.
the currently available oral formulation of CyA. A relative oral bioavail-
ability of 137% in rats compared with Sandimmune Neoral was demon- 7.2. P-gp inhibition by using specic and non-specic P-gp inhibitors
strated for CyA-loaded polymeric micelles. The other aim of the work
was to study the transport mechanism of mPEG-PLA micelles across Lipid excipients and formulations incorporating specic active com-
the intestinal barrier. It was found that polymeric micelles signicantly pounds/inhibitors of P-gp continue to serve as a useful approach to in-
increased the permeability of CyA across Caco-2 monolayers without hibit P-gp and improve the oral bioavailability of drugs with P-gp
signicantly affecting transepithelial electrical resistance values, and limited absorption. Reduction in the bioavailability of PTX due to efux
the apparent permeation coefcient (Papp) of CyA was signicantly by P-gp has been a major challenge in its successful oral delivery. It was
higher in the A B direction compared to that in the B A direction, reported by Woo et al. that in the rat, approximately 54% of the dose of
suggesting that polymeric micelles underwent an active A B transport the orally administered PTX is lost due to efux by P-gp, 38% due to gut
that probably involved endocytosis which was conrmed by confocal lumen metabolism, and 3.5% by gut wall and liver metabolism [38]. Im-
microscope observation. The permeation of CyA through Caco-2 provement of the oral bioavailability of PTX has been widely reported in
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 87

Fig. 8. Various strategies to inhibit/modulate P-gp efux pump, (A) Specic and non-specic P-gp inhibitors, (B) P-gp inhibition by using specic, non-specic and other functional
excipients, (C) P-gp circumvention by using novel peptide prodrug approach.

the literature [48,49,150,151]. The most commonly used approach is the its non-immunosuppressive analog PSC833 [151]. In a study, 14CPTX
co-administration of a specic P-gp substrate and/or incorporation of a in a solution containing Cremophor: ethanol (1:1) that was further di-
nonspecic inhibitor in the formulation. Woo et al. using KR-30031, a luted with saline was administered perorally to anesthetized rats with
verapamil analog with fewer cardiovascular effects than verapamil, and without the co-administration of TPGS (50 mg) or Verapamil
showed that in Caco-2 cells the A B transport of PTX (5 mM) was in- (25 mg). Both TPGS and Verapamil increased the Cmax and AUC of PTX
creased in the presence of KR-30031 (25 mM) and this effect on drug ef- although the effect of TPGS on both parameters was greater. The bio-
ux was similar to that of verapamil (25 mM). A microemulsion availability of PTX increased 6.3-fold and 4.2-fold with TPGS and Verap-
formulation containing dimethylisosorbide, Tween 80 and DL--to- amil, respectively [22]. Chaurasiya et al. investigated the concomitant
copherol and incorporating 6 mg/mL PTX was administered by oral ga- use of SMEDDS and a novel third-generation P-gp inhibitor, GF120918
vage to anesthetized rats. A dose of 25 mg/kg of PTX was used with and (elacridar), for the effective transport of taxanes (PTX and DTX) [25].
without 5,10, 20, and 30 mg/kg of KR-30031 or 20 mg/kg of Verapamil. The investigation was done across an in vitro model of the intestinal ep-
KR-30031 increased the oral bioavailability of PTX from 4.6% to 41% ithelium and uptake into tumour cells. In Caco-2 cell permeation study,
(34% P-gp and 7% liver metabolism inhibition) in a dose dependent PTX-loaded SMEDDS along with GF120918 showed a four-fold increase
manner and this effect was saturable above 20 mg/kg of KR-30031 in apparent permeability, while DTX-loaded SMEDDS in combination
[38]. Earlier studies in improving the oral absorption of PTX were fo- with GF120918 showed a nine-fold increase in permeability, as com-
cused on the co-administration of CyA or one of its analogs. By combin- pared to plain drug solution. Cell uptake studies on A549 cells were per-
ing the marketed lipid formulations of PTX (Taxol) and CyA formed with microemulsions formed from both SMEDDS formulations
(Sandimmune), the peroral bioavailability of PTX in mice increased loaded with rhodamine 123 dye and showed good uptake than plain
from 9.5% to 67%. Increase was 10-fold when CyA was substituted by dye solution. Confocal laser scanning microscopic images further
88 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

conrmed the higher uptake of both SMEDDS formulations in the pres- can be administered orally to HIV (+) patients. Thus transporter
ence of GF120918. targeted prodrug modication of P-gp substrates could lead to shielding
of drug molecules from efux pump. This approach has high clinical sig-
7.3. P-gp circumvention by using novel peptide prodrug approach nicance as it can aid in enhancing intestinal absorption and oral bio-
availability of poorly absorbed PIs and other P-gp substrates.
Besides efux transporters, such as P-gp, a number of nutrient inux
transporters are also expressed on the cellular membranes. These nutri- 8. Current roadblocks in the clinical development of formulation
ent transporters are responsible for the inux of various nutrients and strategies
drugs into various epithelial (enterocytes) and endothelial cells (e.g.
blood brain barrier) [152154]. Recently, peptide transporters (PEPT1 As highlighted in this article, the various formulation strategies for
and PEPT2) have gained lot of attention among drug delivery scientists. BCS Class IV drugs can be explored for their successful delivery. The ben-
Intestinal peptide transporter especially PEPT1, has been a key target ecial properties of various strategies can be taken advantage of in
protein for prodrug design. Several prodrugs have already been de- many ways for the design of impactful medicines and dosage forms. Sig-
signed such that the modied compounds become substrates of peptide nicant expansion of this sector is expected in the upcoming years as
transporters leading to enhanced absorption of these compounds across more and more drugs in the development pipeline are added to the
various physiological barriers [155157]. When a substrate binds to a portfolio of BCS Class IV drugs. However, development of a successful
nutrient transporter it triggers a congurational change in the transport BCS Class IV drug delivery has to face signicant challenges on their
protein as a result of which it is translocated across the membrane and way to the clinics as shown in Table 5.
released into the cell cytoplasm. During this process the substrate is not First and foremost, the bottleneck in the translation of sophisticated
freely available in the inner leaet of the cell membrane and may avoid formulation strategies is the production of large-scale batches under
recognition by P-gp as a substrate. Good Manufacturing Practices (GMP) conditions. The various formulation
Jain et al. have reported that dipeptide conjugates of SQV circumvent strategies discussed in the article are often developed, initially, in the lab
P-gp mediated efux which resulted in enhanced permeability across and there is a signicant gap between academic and industrial production
MDCK-MDR1 cells [158]. Peptide transporters had been exploited in settings. Micrograms or milligrams of product are usually produced in ac-
this study since these transporters are robust and high capacity trans- ademic laboratories while grams or kilograms are necessary for pre-clin-
porters. Utilization of this transporter to ferry the dipeptide prodrugs ical screening, clinical trials, and, ultimately, clinical use. As a general
of SQV across the physiological barriers proved to be a highly effective notion scale up of any laboratory process is difcult; but scaling up nano-
strategy. Similarly, Jain et al. have demonstrated that peptide prodrug particle based delivery systems production is even more challenging
modication of SQV to valine-valine-saquinavir (Val-Val-SQV) and gly- [163165] because subtle variations in the manufacturing procedure
cine-valine-saquinavir (Gly-Val-SQV) can lead to an increased perme- can signicantly alter the product characteristics, which ultimately deter-
ability across rat jejunum [159]. Absorption rate constants in rat mine the therapeutic outcome. In addition, the formulation of drug deliv-
jejunum (ka) for SQV, Val-Val-SQV and Gly-Val-SQV were found to be ery systems is usually a multi-step process. At small scale, it is easy to
14.1 3.4 103, 65.8 4.3 103, and 25.6 5.7 103 min1, achieve reproducibility; however, at the larger, industrial scale, successive
respectively. Enhanced absorption of Val-Val-SQV and Gly-Val-SQV rel- operation streams become more complicated to control making batch-to-
ative to SQV can be attributed to their translocation by the peptide batch reproducibility more difcult.
transporter in the jejunum. Such increased permeability could be attrib- As for any pharmaceutical product, the implementation of a robust
uted partially to the peptide transporter mediated inux of these agents quality control (QC) system is the key to ensure successful manufacturing.
and partially to circumvention of their P-gp mediated efux. Next, Jain Identication of the product's critical quality attributes will help set up
et al. demonstrated that the prodrugs have signicantly lower interac- standards to determine when a batch meets or fails the appropriate re-
tion with MRP-2 relative to SQV [160]. Transepithelial transport of quirements and can be released or not. Incorporating a quality by design
Val-Val-SQV and Gly-Val-SQV across MDCKII-MRP2 cells exhibited an approach upfront in product development will contribute to gain thor-
enhanced absorptive ux and reduced secretory ux as compared to ough product and process knowledge and therefore dene solid QC sys-
SQV. Intestinal perfusion studies revealed that synthesized prodrugs tem [166]. Also, the development of tightly controlled and robust
had higher intestinal permeabilities relative to SQV. They concluded manufacturing should be established as early as possible in the develop-
that peptide transporter targeted prodrug modication of MRP-2 sub- ment process to facilitate the identication of process parameters that
strates lead to shielding of these drug molecules from MRP-2 efux can be deleterious for the product and to ensure batch-to-batch reproduc-
pumps. On the other hand, Agrawal et al. attempted to evade the rst- ibility. This necessitates cooperation between R&D, QC and manufacturing
pass metabolism and efux of LPV by synthesizing peptide prodrugs subdivisions [166]. The process variables can include environmental con-
valinevalinelopinavir (V-V-LPV) and glycinevalinelopinavir (G-V- ditions (temperature, pH, pressure), formulation parameters (character-
LPV) [161]. Both V-V-LPV and G-V-LPV displayed better solubility, istics and origin of raw materials, ratio between the different
metabolism and intestinal permeability proles as compared with the components, solvents used) or be related to the formulation operations
parent drug LPV. Since these prodrugs were substrates for peptide (time, speed, ow conditions) and/or the equipment used. The various
transporters, they bypassed efux pumps and were capable of deliver- formulation strategies discussed in this article usually implements a num-
ing more intact drug to improve oral bioavailability. Evasion of efux ber of operations (e.g., emulsication, sonication, centrifugation, extru-
also lowered the chance of resistance development on chronic LPV ex- sion, solvent evaporation, purication, lyophilization, and/or
posure. Thus, efcient delivery of LPV resulted in enhanced therapeutic sterilization) that all need to be optimized and controlled in series [22,
efcacy which is of high clinical signicance in HAART. Rouquayrol et al. 47]. The use of process optimization software such as Aspen, ProSim can
examined the transepithelial transport of various protease inhibitor help identify the key parameters and optimize the manufacturing design,
(IDV, SQV, and NFV) conjugates using Caco-2 cell monolayers as even at an early stage of the development. In addition, quality by design
a model of the intestinal barrier [162]. They found that conjugation of and extensive formulation testing under a variety of conditions can help
L-valine, L-leucine or L-phenylalanine (through their carboxyl) to the construct a comprehensive understanding of relations between
protease inhibitors constitutes a most appealing alternative, which im- manufacturing conditions and nal product characteristics. The denition
proves their absorptive diffusion and reduces their recognition by efux of critical steps and variables will in turn facilitate scaling up the
carriers. The 3- to 6-fold absorption enhancement resulting from the manufacturing process [167169].
conjugation of the protease inhibitors to the amino acids indicates In addition, adequate analytical methods have to be developed and
that potentially lower doses of protease inhibitors (as their conjugates) validated to assess product quality at the intermediate and nal steps.
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 89

For example, lipid and nanoparticle based delivery systems require a mechanism, and their manufacture should be provided to persuade
panel of analytics that are specic to this class of systems and unusual the regulatory authorities of their acceptability [177]. Safety assessment
in classical pharmaceutical development. The literature on their and the potential inuence of biopharmaceutical factors on the drug or
manufacturing processes and industrial characterization methodologies lipid excipients need to be explored. It may be difcult to predict in vivo
is limited [170172]. Nevertheless, some of the specic characterization performances of a lipid dosage form based on in vitro results obtained
methods common to lipid and nanoparticle-based drug delivery systems through conventional dissolution methods in view of the convoluted
include dynamic light scattering (DLS) for size and size distribution mea- GI processing of lipid formulations. More mechanistic studies should
surement, Zetasizer (Malvern) for zeta potential measurement, microsco- be conducted to facilitate a better understanding of the pharmaceutical
py techniques (AFM, SEM, cryo-TEM) as a complementary size characteristics of lipid formulations and interactions between lipid ex-
determination method and/or for morphology. As the volume of cipients, drug, and physiological environment. The lack of predictability
manufacturing grows, there will need to be further discussion among for product quality and performance may be due to the nature of empir-
the eld as to what tools or techniques are most appropriate for each ical and iterative processes traditionally employed [178].
physico-chemical property, which software or data processing method With the aim of rationalizing the design of lipid formulation and to
is most relevant to handle these data sets, and how these data should better understand the fate of a drug after oral administration in a
be reported and communicated. In addition, these methods have to be lipid-based formulation, a consortium, composed of academics and in-
sensitive enough to detect potential small variations that could impact dustrial scientists, has been created (http://www.lfcsconsortium.org/).
the nal product quality and a major challenge will be the implementa- The consortium sponsors and conducts research to develop in vitro
tion of in-process controls along the manufacturing chain. Also, training methods to assess the performance of LBDDS during dispersion and di-
the personnel to the specicities, hurdles and challenges related to the gestion, which are critical parameters. The primary objective is to devel-
discussed formulation strategies is an important aspect of the success of op guidelines that rationalize and accelerate the development of drug
the manufacturing. formulations through the identication of key performance criteria
Furthermore, for systems that are intended for parenteral adminis- and the validation and eventual publication of universal standard tests
tration, nding an adequate method of sterilization is a nal crucial and operating procedures [179]. In order to establish approved guide-
step of the manufacturing, and has to be validated on a case by case lines, appropriate dialogue with pharmaceutical regulatory bodies
basis, as the formulation can be impacted differently by the sterilization (FDA, EMEA) is also foreseen.
method, depending on their components, formulation strategy and/or More consideration needs to be paid to the characteristics of various
method of preparation [172,173]. Gamma irradiation or autoclaving lipid formulations available, so that guidelines and experimental
can be damaging for certain formulation strategies and can affect prod- methods can be established that allow identication of candidate for-
uct characteristics. Sterile ltration of the nal product can be an alter- mulations at an early stage. Methods need to be sought for tracking
native for systems with a size distribution below 0.22 m. If the size is the solubilization state of the drug in vivo, and there is a need for in
greater than the lter cut-off, loss of material might occur. Finally, asep- vitro methods for predicting the dynamic changes, which are expected
tic technique is another possible option to consider, even though it can to take place in the gut. Attention to the physical and chemical stability
be difcult to execute in the case of multi-steps processes [171,173]. of drugs within lipid systems and the interactions of lipid systems with
Finally, it should be noted that most of the development of nanoparti- the components of capsule shells will also be required. Whilst these
cle-based delivery systems and investigational strategies involving the P- present challenges there is a great potential in the use of lipid formula-
gp inhibitors is still done in academic labs, start-ups or small and medium tions. The priority for future research should be to conduct human bio-
enterprises (SME). The last two represent 75% of the 200 companies de- availability studies and to conduct more basic studies on the
veloping nanoparticle-based products. For these small entities, securing mechanisms of action of this fascinating and diverse group of
investments is crucial to product development, which is a long and formulations.
cost-intensive process. In addition to the technical and manufacturing Pharmaceutical co-crystals have not yet been ofcially addressed
challenges, they have to face the traditional reluctance of large pharma- from the perspective of generic regulatory approval. In some respects,
ceutical companies to nanoproducts and often have to provide stronger co-crystals are intermediate between hydrates/polymorphs (which
preclinical data sets compared to small molecules on account of the are ANDA-eligible) and salts (which, in general, are not). Unlike poly-
lower perceived chance of success and the technical complexity that can morphs, which generally speaking contain only the API within the crys-
discourage investors or large pharmaceutical companies to move for- tal lattice, co-crystals are composed of an API with a neutral guest
ward. The high cost associated with nanoparticle-based product develop- compound (also referred to as a conformer) in the crystal lattice [180].
ment favours the propensity toward specic therapeutic indications, such Similarly, unlike salts, where the components in the crystal lattice are
as oncology where the market is huge and rare diseases where the regu- in an ionized state, a co-crystal's components are in a neutral state
latory barrier to entry is lower [174176]. and interact via non-ionic interactions. Like hydrates, co-crystals are
non-ionic supramolecular complexes; but like salts, co-crystals involve
9. Regulatory aspects for the development of BCS class IV delivery complexation with substances of greater potential toxicity than water
systems [181]. At present no formal regulatory policy exists governing the clas-
sication of pharmaceutical co-crystals. In response to the need for reg-
Several drug delivery systems of BCS Class IV drugs have been ap- ulatory guidance, the Ofce of Pharmaceutical Science Centre for Drug
proved by the US Food and Drug Administration (FDA) and European Evaluation and Research (CDER) at the FDA has issued guidance for
Medicines Agency (EMA) for a variety of therapeutic indications as pharmaceutical co-crystals [182]. This guidance provides the current
shown in Table 4. Signicant expansion of this sector is expected in up- thinking on the appropriate classication of co-crystal solid-state
coming years however, the developers have to face signicant regulato- forms, the data that should be submitted to support the classication,
ry challenges on their way to the clinics. and the regulatory implications of such a classication.
From a regulatory point of view, quality and safety issues associated Similarly, for nanoparticle-based delivery systems there are current-
with preclinical and clinical studies are the key difculties likely to be ly no specic requirements from the regulatory agencies (FDA and
encountered in launching a lipid-based dosage form in the market, EMA) for the preclinical and clinical testing, only reection papers pro-
and above all the manifestations of the therapeutic efcacy. The overall viding guidelines on the pharmaceutical development of a specic type
drug stability and absence of immunological reactions to the oils or lipid of nanoparticle-based drug delivery systems have been published [183],
excipients have to be demonstrated. Sufcient details explaining the use and to date the evaluation process follows a similar path as for small-
of lipid excipients and the types of dosage form, the drug release molecules drugs. The development of a new drug starts with preclinical
90 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

Table 4
FDA and EMA approved BCS Class IV drug products and their therapeutic indications.

Authorized
BCS class IV drug Brand Company Formulation strategy markets Indication

Cyclosporine Neoral Novartis SMEDDS US Organ transplantation, rheumatoid arthritis, psoriasis


Sandimmune Novartis SMEDDS US Organ transplantation
Gengraf Abbott SMEDDS US Organ transplantation
Panimun Bioral Panacea Biotec SMEDDS US Organ transplantation
Restatis Allergan Anionic microemulsion US Keratoconjunctivitis sicca (dry eye syndrome)
Ikervis Santen Pharmaceuticals Cationic microemulsion Europe Keratoconjunctivitis sicca (dry eye syndrome)
Aprepitant Emend Merck & Co. Nanocrystals US, Europe Chemotherapy induced nausea and vomiting
(suspension)
Saquinavir Fortavase Hoffmann La Roche, Inc. SMEDDS US HIV infection
Invirase Hoffmann La Roche, Inc. SMEDDS US HIV infection
Ritonavir Norvir Abbott SMEDDS US HIV infection
Ritonavir; Kaletra Abbott SMEDDS US HIV infection
Lopinavir
Amphotericin B AmBisome Astellas Liposome US Fungal infections, leishmaniasis
Abelcet Sigma-tau Lipid complex US Fungal infections
Paclitaxel Taxol HQ Speciality Pharma Micelles US Breast cancer, lung cancer, ovarian cancer
Abraxane Abraxis BioScience Albumin nanoparticles US, Europe Mestatic breast cancer
Docetaxel Taxotere Sano aventis Micelles US, Europe Breast cancer, prostate cancer, non-small cell lung cancer
Docefrez Sun Pharma Micelles US, Europe Breast cancer, prostate cancer, non-small cell lung cancer

*The list provided above is restricted to the formulation strategies discussed in this article and does not take into account the conventional dosage forms of BCS Class IV drugs approved by
FDA and EMA.

testing followed by the submission of an Investigational New Drug opportunity to clarify the manufacturer's obligations and discuss
(IND) application in order to initiate the clinical trial, which is organized methodologies and data needed to meet those obligations [186].
in three phases (Phase I, II and III clinical trials). Only for approximately Not all excipients are inert substances, and some may be toxic at
5% of the initial tested entities (including all INDs, nanoparticle based augmented concentrations. In the Code of Federal Regulations, the
and non-nanoparticle based) will the clinical evaluation procedure re- FDA has published a list of substances that are generally recognized as
sult in the submission of a New Drug Application (NDA) and, ultimately, safe (GRAS). Apart from this, it also maintains a list of inactive ingredi-
market authorization. Each new nanoparticle-based drug delivery sys- ents for excipients entitled inactive ingredient database (IID) that are
tem has to follow the complete evaluation process, even if the active approved and can be incorporated in marketed products. This guide
pharmaceutical ingredient (API) alone has been in clinical use for provides the list of maximum amount allowed for excipients, which
many years [176]. The pharmaceutical regulatory environment, the can be used for a specic route of administration. Once an inactive in-
health care strategies, the demographics and the wide economic envi- gredient has been approved for a product through a particular route of
ronment affect the competitive nanomedicine-related drug market ap- administration, it can be used in any new drug formulation and does
proval. Despite all difculties along the development process, a not require extensive review. The formulator can take the information
considerable number of nanoparticle-based drug delivery systems are from both GRAS and IIG when developing a new formulation. Currently,
already in the market, approved by the FDA, EMA or foreign equivalent the FDA does not have any process or mechanism to evaluate the safety
[184]. of excipients individually. Instead, the excipients are reviewed and ap-
The FDA recognizes that nanotechnology is an emerging technol- proved as components of the drug or biological product in the applica-
ogy that has the potential to be used across the full spectrum of FDA- tion. Since excipients play an integral part in the formulation and cannot
regulated products, including drugs, biological products, and medi- be reviewed separately from the drug formulation, the regulatory pro-
cal devices. Over the past several years, the FDA has taken multiple cess is appropriate from a scientic standpoint [179].
steps to help ensure the responsible development of nanotechnology
products. In 2011, the FDA issued a draft guidance for industry enti- 10. Conclusion
tled Considering Whether an FDA regulated Product Involves the
Application of Nanotechnology to present its current thinking on The formulation of drugs is carried out with the principle objective of
nanotechnology and to seek public comment [185,186]. As noted in enhancing their bioavailability. The BCS class IV drugs which exhibit
that draft guidance, the FDA has not adopted a regulatory denition poor solubility and membrane permeability are challenging for the for-
of nanotechnology or related terms. The FDA's Centre for Drug Eval- mulation scientist with regard to bioavailability and exhibiting opti-
uation and Research provided an update on its ongoing work related mum therapeutic potential. The various formulation development
to the use of nanotechnology in drug products to the Advisory Com- strategies highlighted in this review like lipid based delivery systems,
mittee for Pharmaceutical Science and Clinical Pharmacology in Au- polymer based nanocarriers, crystal engineering (nanocrystals and co-
gust 2012 [187]. As explained in this update, the FDA evaluated its crystals), liquisolid technology, self-emulsifying solid dispersions can
existing regulatory review processes for drug products and deter- be explored and experimented further to increase their commercial ap-
mined that current procedures are adequate to identify and address plicability. For example, the recent advances in LDBBS have led to the
the potential risks associated with the use of nanomaterials in drug successful commercialization of few BCS class IV [saquinavir
products. Certain areas were identied for improvement, including (Fortovase), ritonavir (Norvir)] lipid-based formulations. Similarly,
the need for increased regulatory science research and the training the advent of novel scalable technologies for the production of
of review staff. The FDA also established specic policies and proce- nanocrystals has eased the journey of BCS Class IV drug aprepitant
dures for use by internal reviewers of human drug and animal drug (Emend) from researcher's bench side to clinic. The progressive eluci-
submissions. As noted in the draft guidance documents, the FDA en- dation of the remaining strategies and their mechanisms involved in
courages industry to consult early with the agency to address ques- improving the bioavailability of BCS Class IV drugs is yet to be addressed
tions related to the safety, effectiveness, or regulatory status of in depth to advance the technology. Further research has to be carried
nanotechnology products. These early consultations afford an out regarding the design of a proper in vivo model to correlate the
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 91

Table 5
The advantages and bottlenecks to commercialization of BCS Class IV drug formulation strategies.

Formulation
strategy Advantages Bottlenecks to commercialization

Lipid based Proven commercial and pharmaceutical benet for compounds Insufcient drug solubility in the excipient matrix, which precludes administration
drug delivery such as CyA (Sandimmune) and the HIV protease inhibitors of the entire dose in a single oral capsule
systems (Kaletra) Physical and chemical stability of drugs solubilized in lipid excipients have not yet
Ability to encapsulate high drug content (compared to other car- been adequately addressed
rier systems e.g. polymeric nanoparticles) Lipid excipients themselves may be subject to physical changes or chemical degra-
The feasibility of carrying both lipophilic and hydrophilic drugs dation over time
Most of the lipids used are biodegradable, and as such they have The excipients need to be used in their regulatorily accepted concentrations. If
excellent biocompatibility, are non-toxic, non-allergenic and distinctly higher concentrations need to be used, a limited toxicity study should be
non-irritating performed to prove the safety of the excipients at that concentration.
Formulated by water-based technologies and thus can avoid or-
ganic solvents
Easy to scale-up and sterilize and are less expensive than
polymeric/surfactant based carriers
Easy to validate.
Polymeric Proven commercial and pharmaceutical benets for a few formu- Lack of standard nano nomenclature: imprecise denition for nanomedicines
nanocarriers lation strategies like polymeric micelles (Taxotere, Taxol) Lack of precise control over nanoparticle manufacturing parameters and control
Increased solubilization potential and superior encapsulation assays
Chemical versatility of materials eligible for nanomedicines Currently used compounds/components for synthesis of nanoparticle based deliv-
Versatile routes of administration ery systems often pose problems for large scale good manufacturing (cGMP) pro-
duction
Lack of quality control: issues pertaining to separation of undesired nanostructures
(byproducts, catalysts, starting materials) during manufacturing
Scalability complexities: enhancing the production rate to increase yield
Reproducibility issues: control of particle size distribution and mass
High fabrication costs
Lack of rational pre-clinical characterization strategies via multiple techniques
Biocompatibility, bio distribution and toxicity issues: lack of knowledge regarding
the interaction between nanoparticles and bio surfaces/tissues
Consumer condence: public's general reluctance to embrace innovative medical
technologies without clearer safety or regulatory guidelines
Relative scarcity of venture funds
Ethical issues and societal issues are hyped up by the media
Big pharma's continued reluctance to seriously invest in nanomedicine
Patent review delays, patent thickets, and issuance of invalid patents by the U.S.
Patent and Trademark Ofce
Regulatory uncertainty and confusion due to baby steps undertaken by U.S. Food
and Drug Administration: a lack of clear regulatory/safety guidelines
P-gp inhibition Proven commercial and pharmaceutical benets for formulation Highly experimental approach limited to academic lab settings and yet to be ex-
approach strategy dealing with co-administration of P-gp inhibitor plored
(Kaletra) Biocompatibility, toxicity data related to various P-gp inhibitors is limited
Versatile routes of administration Scalability complexities: enhancing the production rate to increase yield
High fabrication cost
Self-emulsifying Availability of newer thermo-softening surface active and self-- Physical and chemical stability of drugs solubilized in lipid excipients have not yet
solid emulsifying excipients been adequately addressed
dispersions Can be lled directly into hard gelatin capsules in molten state, Lipid excipients themselves may be subject to physical changes or chemical degra-
obviating the requirement of milling and blending prior to lling dation over time
Industrially feasible manufacturing techniques available like hot
melt extrusion
Liquisolid Industrially feasible manufacturing techniques available Regulatory scenario (NDA application and review process) is still obscure and the
technology regulatory agencies need to shed some light on it
Nanocrystals Proven commercial and pharmaceutical benets for aprepitant Maintenance of particle size, size distribution and polymorphism poses a particu-
(Emend) larly big challenge
Versatile routes of administration Strategic control of crystal morphology and shape is by and large an expertise of
Industrially feasible manufacturing techniques available precipitation techniques (bottom up approaches) and is a face of research which is
Adept to convenient sterilization techniques a key driver for de- yet to be explored effectively
velopment of parenteral formulations The lab scale equipment presents higher surface area to volume ratios while the
Incorporation of appropriate stabilizers followed by drying production level equipments presents a higher volume to surface ratio. This differ-
(freeze/spray) can ensure long term stability ence is one of the considerable factors which slows down the processing during
Opportunity to produce generic versions of presently available scale up.
costly drugs Large scale equipment consumes more time when compared to lab equipment.
These long processing times are likely to lead to adverse conditions such as
adsorption, precipitation, viscosity alterations and may affect the mass and mo-
mentum transfer rates, the shear rate and stress factors
Co-crystals Creates intellectual property for a new patent of an API for ex- Regulatory scenario (NDA application and review process) is still obscure and the
tending their life cycle regulatory agencies need to shed some light on it
Versatile routes of administration

data obtained in vitro studies to the actual in vivo experience. In conclu- for these highly notorious BCS class IV drugs. Also, before commerciali-
sion, a number of delivery options have been developed for improving zation of the various formulations strategies, it will be essential to per-
the physicochemical and pharmacokinetic behaviours of BCS Class IV form pharmacoeconomic studies to demonstrate both social and
drugs in academic and industrial research. A better understanding of economic added value of these novel products when compared with
the physicochemical properties of drug substances and the limitations established treatments. Also, in case of formulations pertaining to anti-
of each delivery option would lead to efcient formulation development cancer agents the important indicators such as increment in QALY
92 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

(quality-adjusted life expectancy years) or costs associated to future [26] D.J. Patel, J.K. Patel, Design and evaluation of famotidine mucoadhesive nanoparti-
cles for aspirin induced ulcer treatment, Braz. Arch. Biol. Technol. 56 (2013)
consecutive hospitalizations should be considered in the development 223236.
of these new sophisticated formulations. From a regulatory perspective [27] J.S. Negi, P. Chattopadhyay, A.K. Sharma, V. Ram, Development and evaluation of
also, the scenario is obscure and the various regulatory agencies need to glyceryl behenate based solid lipid nanoparticles (SLNs) using hot self-
nanoemulsication (SNE) technique, Arch. Pharm. Res. 37 (2014) 361370.
throw light on the various guidelines to be adhered, for the successful [28] M. Schller-Gyre, T.N. Kakuda, A. Raoof, G. De Smedt, R.M. Hoetelmans, Clinical
commercialization of the novel formulation strategies. pharmacokinetics and pharmacodynamics of etravirine, Clin. Pharmacokinet. 48
(2009) 561574.
[29] S.S. Rane, B.D. Anderson, What determines drug solubility in lipid vehicles: is it
References predictable? Adv. Drug Deliv. Rev. 60 (2008) 638656.
[30] S. Chakraborty, D. Shukla, B. Mishra, S. Singh, Lipidan emerging platform for oral
[1] X.Y. Lawrence, E. Lipka, J.R. Crison, G.L. Amidon, Transport approaches to the bio- delivery of drugs with poor bioavailability, Eur. J. Pharm. Biopharm. 73 (2009)
pharmaceutical design of oral drug delivery systems: prediction of intestinal ab- 115.
sorption, Adv. Drug Deliv. Rev. 19 (1996) 359376. [31] C.J. Porter, W.N. Charman, Uptake of drugs into the intestinal lymphatics after oral
[2] D. Sun, L.X. Yu, M.A. Hussain, D.A. Wall, R.L. Smith, G.L. Amidon, In vitro testing of administration, Adv. Drug Deliv. Rev. 25 (1997) 7189.
drug absorption for drug'developability'assessment: forming an interface between [32] E. Roger, F. Lagarce, E. Garcion, J.-P. Benoit, Lipid nanocarriers improve paclitaxel
in vitro preclinical data and clinical outcome, Curr. Opin. Drug Discov. Dev. 7 transport throughout human intestinal epithelial cells by using vesicle-mediated
(2004) 7585. transcytosis, J. Control. Release 140 (2009) 174181.
[3] G.L. Amidon, H. Lennerns, V.P. Shah, J.R. Crison, A theoretical basis for a [33] G. Cornaire, J. Woodley, P. Hermann, A. Cloarec, C.C. Arellano, G. Houin, Impact of
biopharmaceutic drug classication: the correlation of in vitro drug product disso- excipients on the absorption of P-glycoprotein substrates in vitro and in vivo, Int.
lution and in vivo bioavailability, Pharm. Res. 12 (1995) 413420. J. Pharm. 278 (2004) 119131.
[4] C.A. Lipinski, F. Lombardo, B.W. Dominy, P.J. Feeney, Experimental and computa- [34] Y. Shono, H. Nishihara, Y. Matsuda, S. Furukawa, N. Okada, T. Fujita, A. Yamamoto,
tional approaches to estimate solubility and permeability in drug discovery and Modulation of intestinal P-glycoprotein function by cremophor EL and other sur-
development settings, Adv. Drug Deliv. Rev. 64 (2012) 417. factants by an in vitro diffusion chamber method using the isolated rat intestinal
[5] C. Giliyar, D.T. Fikstad, S. Tyavanagimatt, Challenges and opportunities in oral de- membranes, J. Pharm. Sci. 93 (2004) 877885.
livery of poorly water-soluble drugs, Drug Deliv. Technol. 6 (2006) 5763. [35] M.M. Nerurkar, P.S. Burton, R.T. Borchardt, The use of surfactants to enhance the
[6] P. Stenberg, C.A.S. Bergstrm, K. Luthman, P. Artursson, Theoretical predictions of permeability of peptides through Caco-2 cells by inhibition of an apically polarized
drug absorption in drug discovery and development, Clin. Pharmacokinet. 41 efux system, Pharm. Res. 13 (1996) 528534.
(2002) 877899. [36] V.J. Wacher, S. Wong, H.T. Wong, Peppermint oil enhances cyclosporine oral bio-
[7] P.-L. Toutain, A. Bousquet-MLou, Bioavailability and its assessment, J. Vet. availability in rats: comparison with D-alpha-tocopheryl poly (ethylene glycol
Pharmacol. Ther. 27 (2004) 455466. 1000) succinate (TPGS) and ketoconazole, J. Pharm. Sci. 91 (2002) 7790.
[8] M. Lindenberg, S. Kopp, J.B. Dressman, Classication of orally administered drugs [37] N. Anton, T.F. Vandamme, Nano-emulsions and micro-emulsions: clarications of
on the World Health Organization Model list of Essential Medicines according to the critical differences, Pharm. Res. 28 (2011) 978985.
the biopharmaceutics classication system, Eur. J. Pharm. Biopharm. 58 (2004) [38] J.S. Woo, C.H. Lee, C.K. Shim, S.-J. Hwang, Enhanced oral bioavailability of paclitaxel
265278. by coadministration of the P-glycoprotein inhibitor KR30031, Pharm. Res. 20
[9] L. Perioli, V. Ambrogi, M. Nocchetti, M. Sisani, C. Pagano, Preformulation studies on (2003) 2430.
hostguest composites for oral administration of BCS class IV drugs: HTlc and furo- [39] Y.-M. Yin, F.-D. Cui, C.-F. Mu, M.-K. Choi, J.S. Kim, S.-J. Chung, C.-K. Shim, D.-D. Kim,
semide, Appl. Clay Sci. 53 (2011) 696703. Docetaxel microemulsion for enhanced oral bioavailability: preparation and in
[10] A.M. Kaukonen, L. Laitinen, J. Salonen, J. Tuura, T. Heikkil, T. Limnell, J. Hirvonen, vitro and in vivo evaluation, J. Control. Release 140 (2009) 8694.
V.-P. Lehto, Enhanced in vitro permeation of furosemide loaded into thermally car- [40] D. Butani, C. Yewale, A. Misra, Amphotericin B topical microemulsion: formulation,
bonized mesoporous silicon (TCPSi) microparticles, Eur. J. Pharm. Biopharm. 66 characterization and evaluation, Colloids Surf. B: Biointerfaces 116 (2014)
(2007) 348356. 351358.
[11] P. Fasinu, V. Pillay, V.M. Ndesendo, L.C. du Toit, Y.E. Choonara, Diverse approaches [41] S.K. Jha, R. Karki, D. Venkatesh, A. Geethalakshami, Formulation development & char-
for the enhancement of oral drug bioavailability, Biopharm. Drug Dispos. 32 (2011) acterization of microemulsion drug delivery systems containing antiulcer drug, Int. J.
185209. Drug Dev. Res. (2011).
[12] A. Zvonar, K. Berginc, A. Kristl, M. Gaperlin, Microencapsulation of self- [42] H. Reiss, Entropy-induced dispersion of bulk liquids, J. Colloid Interface Sci. 53 (1975)
microemulsifying system: improving solubility and permeability of furosemide, 6170.
Int. J. Pharm. 388 (2010) 151158. [43] R.N. Gursoy, S. Benita, Self-emulsifying drug delivery systems (SEDDS) for improved
[13] I. Vural, C. Sarisozen, S.S. Olmez, Chitosan coated furosemide liposomes for im- oral delivery of lipophilic drugs, Biomed. Pharmacother. 58 (2004) 173182.
proved bioavailability, J. Biomed. Nanotechnol. 7 (2011) 426430. [44] Y.-D. Yan, N. Marasini, Y.K. Choi, J.O. Kim, J.S. Woo, C.S. Yong, H.G. Choi, Effect of
[14] A. Singh, Z.A. Worku, G. Van den Mooter, Oral formulation strategies to improve sol- dose and dosage interval on the oral bioavailability of docetaxel in combination
ubility of poorly water-soluble drugs, Expert Opin. Drug Deliv. 8 (2011) 13611378. with a curcumin self-emulsifying drug delivery system (SEDDS), Eur. J. Drug
[15] A. Beloqui, M.. Solins, A.R. Gascn, A. del Pozo-Rodrguez, A. des Rieux, V. Prat, Metab. Pharmacokinet. 37 (2012) 217224.
Mechanism of transport of saquinavir-loaded nanostructured lipid carriers across [45] A.A. Sultan, S.A. El-Gizawy, M.A. Osman, G.M. El Maghraby, Colloidal carriers for ex-
the intestinal barrier, J. Control. Release 166 (2013) 115123. tended absorption window of furosemide, J. Pharm. Pharmacol. (2016).
[16] N.A. Kasim, M. Whitehouse, C. Ramachandran, M. Bermejo, H. Lennerns, A.S. [46] Y.G. Seo, D.H. Kim, T. Ramasamy, J.H. Kim, N. Marasini, Y.-K. Oh, D.-W. Kim, J.K. Kim,
Hussain, H.E. Junginger, S.A. Stavchansky, K.K. Midha, V.P. Shah, Molecular proper- C.S. Yong, J.O. Kim, Development of docetaxel-loaded solid self-nanoemulsifying
ties of WHO essential drugs and provisional biopharmaceutical classication, Mol. drug delivery system (SNEDDS) for enhanced chemotherapeutic effect, Int. J.
Pharm. 1 (2004) 8596. Pharm. 452 (2013) 412420.
[17] M.E. Taub, L. Kristensen, S. Frokjaer, Optimized conditions for MDCK permeability [47] Y. Chen, C. Chen, J. Zheng, Z. Chen, Q. Shi, H. Liu, Development of a solid
and turbidimetric solubility studies using compounds representative of BCS classes supersaturatable self-emulsifying drug delivery system of docetaxel with im-
IIV, Eur. J. Pharm. Sci. 15 (2002) 331340. proved dissolution and bioavailability, Biol. Pharm. Bull. 34 (2011) 278286.
[18] F. Ibrahim, P. Gershkovich, O. Sivak, E.K. Wasan, K.M. Wasan, Assessment of novel [48] P. Gao, B.D. Rush, W.P. Pfund, T. Huang, J.M. Bauer, W. Morozowich, M.S. Kuo, M.J.
oral lipid-based formulations of amphotericin B using an in vitro lipolysis model, Hageman, Development of a supersaturable SEDDS (S-SEDDS) formulation of pac-
Eur. J. Pharm. Sci. 46 (2012) 323328. litaxel with improved oral bioavailability, J. Pharm. Sci. 92 (2003) 23862398.
[19] C.-Y. Wu, L.Z. Benet, Predicting drug disposition via application of BCS: transport/ [49] S. Yang, R.N. Gursoy, G. Lambert, S. Benita, Enhanced oral absorption of paclitaxel
absorption/elimination interplay and development of a biopharmaceutics drug in a novel self-microemulsifying drug delivery system with or without concomi-
disposition classication system, Pharm. Res. 22 (2005) 1123. tant use of P-glycoprotein inhibitors, Pharm. Res. 21 (2004) 261270.
[20] P. Sanphui, L. Rajput, Tuning solubility and stability of hydrochlorothiazide co-crys- [50] A. Deshmukh, S. Kulkarni, Solid self-microemulsifying drug delivery system of rito-
tals, Acta Crystallogr. Sect. B: Struct. Sci. Cryst. Eng. Mater. 70 (2014) 8190. navir, Drug Dev. Ind. Pharm. 40 (2014) 477487.
[21] K. Wang, J. Qi, T. Weng, Z. Tian, Y. Lu, K. Hu, Z. Yin, W. Wu, Enhancement of oral [51] B. Garg, O. Katare, S. Beg, S. Lohan, B. Singh, Systematic development of solid self-
bioavailability of cyclosporine A: comparison of various nanoscale drug-delivery nanoemulsifying oily formulations (S-SNEOFs) for enhancing the oral bioavailabil-
systems, Int. J. Nanomedicine 28 (2014) 9. ity and intestinal lymphatic uptake of lopinavir, Colloids Surf. B: Biointerfaces 141
[22] M.V. Varma, R. Panchagnula, Enhanced oral paclitaxel absorption with vitamin E- (2016) 611622.
TPGS: effect on solubility and permeability in vitro, in situ and in vivo, Eur. J. [52] C. Jaafar-Maalej, R. Diab, V. Andrieu, A. Elaissari, H. Fessi, Ethanol injection method
Pharm. Sci. 25 (2005) 445453. for hydrophilic and lipophilic drug-loaded liposome preparation, J. Liposome Res.
[23] J. Moes, S. Koolen, A. Huitema, J. Schellens, J. Beijnen, B. Nuijen, Development of an 20 (2010) 228243.
oral solid dispersion formulation for use in low-dose metronomic chemotherapy of [53] T. Nii, F. Ishii, Encapsulation efciency of water-soluble and insoluble drugs in lipo-
paclitaxel, Eur. J. Pharm. Biopharm. 83 (2013) 8794. somes prepared by the microencapsulation vesicle method, Int. J. Pharm. 298
[24] D.M. Stevens, K.A. Gilmore, E. Harth, An assessment of nanosponges for intrave- (2005) 198205.
nous and oral drug delivery of BCS class IV drugs: drug delivery kinetics and solu- [54] A. Rathore, A. Jain, A. Gulbake, S. Shilpi, P. Khare, A. Jain, S.K. Jain, Mannosylated li-
bilization, Polym. Chem. 5 (2014) 35513554. posomes bearing amphotericin B for effective management of visceral leishmania-
[25] A. Chaurasiya, A.K. Singh, G.K. Jain, M.H. Warsi, E. Sublet, F.J. Ahmad, G. Borchard, sis, J. Liposome Res. 21 (2011) 333340.
R.K. Khar, Dual approach utilizing self microemulsifying technique and novel P- [55] J. Wu, Q. Liu, R.J. Lee, A folate receptor-targeted liposomal formulation for paclitax-
gp inhibitor for effective delivery of taxanes, J. Microencapsul. 29 (2012) 583595. el, Int. J. Pharm. 316 (2006) 148153.
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 93

[56] W. Mehnert, K. Mder, Solid lipid nanoparticles: production, characterization and [88] Y. Mo, L.-Y. Lim, Preparation and in vitro anticancer activity of wheat germ agglu-
applications, Adv. Drug Deliv. Rev. 47 (2001) 165196. tinin (WGA)-conjugated PLGA nanoparticles loaded with paclitaxel and isopropyl
[57] M.A. Alex, A. Chacko, S. Jose, E. Souto, Lopinavir loaded solid lipid nanoparticles myristate, J. Control. Release 107 (2005) 3042.
(SLN) for intestinal lymphatic targeting, Eur. J. Pharm. Sci. 42 (2011) 1118. [89] V. Bhardwaj, J. Plumb, J. Cassidy, M.R. Kumar, Evaluating the potential of polymer
[58] A. Alex, W. Paul, A. Chacko, C.P. Sharma, Enhanced delivery of lopinavir to the CNS nanoparticles for oral delivery of paclitaxel in drug-resistant cancer, Cancer
using Compritol-based solid lipid nanoparticles, Ther. Deliv. 2 (2011) 2535. Nanotechnol. 1 (2010) 2934.
[59] P.R. Ravi, R. Vats, V. Dalal, A.N. Murthy, A hybrid design to optimize preparation of [90] M. Nahar, N.K. Jain, Preparation, characterization and evaluation of targeting po-
lopinavir loaded solid lipid nanoparticles and comparative pharmacokinetic evalu- tential of amphotericin B-loaded engineered PLGA nanoparticles, Pharm. Res. 26
ation with marketed lopinavir/ritonavir coformulation, J. Pharm. Pharmacol. 66 (2009) 25882598.
(2014) 912926. [91] P.R. Ravi, R. Vats, J. Balija, S.P.N. Adapa, N. Aditya, Modied pullulan nanoparticles
[60] A. Beloqui, M.. Solins, A. des Rieux, V. Prat, A. Rodrguez-Gascn, Dextranprot- for oral delivery of lopinavir: formulation and pharmacokinetic evaluation,
amine coated nanostructured lipid carriers as mucus-penetrating nanoparticles for Carbohydr. Polym. 110 (2014) 320328.
lipophilic drugs, Int. J. Pharm. 468 (2014) 105111. [92] P.R. Ravi, R. Vats, V. Dalal, N. Gadekar, Design, optimization and evaluation of poly-
[61] K. Hadinoto, A. Sundaresan, W.S. Cheow, Lipidpolymer hybrid nanoparticles as a -caprolactone (PCL) based polymeric nanoparticles for oral delivery of lopinavir,
new generation therapeutic delivery platform: a review, Eur. J. Pharm. Biopharm. Drug Dev. Ind. Pharm. 41 (2015) 131140.
85 (2013) 427443. [93] J. Italia, M. Yahya, D. Singh, M.R. Kumar, Biodegradable nanoparticles improve oral
[62] Y. Liu, K. Li, J. Pan, B. Liu, S.-S. Feng, Folic acid conjugated nanoparticles of mixed bioavailability of amphotericin B and show reduced nephrotoxicity compared to
lipid monolayer shell and biodegradable polymer core for targeted delivery of Do- intravenous Fungizone, Pharm. Res. 26 (2009) 13241331.
cetaxel, Biomaterials 31 (2010) 330338. [94] H. Van de Ven, C. Paulussen, P. Feijens, A. Matheeussen, P. Rombaut, P. Kayaert, G. Van
[63] S. Jain, P.U. Valvi, N.K. Swarnakar, K. Thanki, Gelatin coated hybrid lipid nanoparti- den Mooter, W. Weyenberg, P. Cos, L. Maes, PLGA nanoparticles and nanosuspensions
cles for oral delivery of amphotericin B, Mol. Pharm. 9 (2012) 25422553. with amphotericin B: potent in vitro and in vivo alternatives to Fungizone and
[64] S. Sambaraj, D. Ammula, V. Nagabandi, Furosemide loaded silica-lipid hybrid mi- AmBisome, J. Control. Release 161 (2012) 795803.
croparticles: formulation development, in vitro and ex vivo evaluation, Adv. [95] J. Italia, D. Bhatt, V. Bhardwaj, K. Tikoo, M.R. Kumar, PLGA nanoparticles for oral deliv-
Pharm. Bull. 5 (2015) 403. ery of cyclosporine: nephrotoxicity and pharmacokinetic studies in comparison to
[65] N.T. Huynh, C. Passirani, P. Saulnier, J.-P. Benoit, Lipid nanocapsules: a new plat- Sandimmune Neoral, J. Control. Release 119 (2007) 197206.
form for nanomedicine, Int. J. Pharm. 379 (2009) 201209. [96] R. Singh, P. Kesharwani, N.K. Mehra, S. Singh, S. Banerjee, N. Jain, Develop-
[66] E. Roger, F. Lagarce, J.-P. Benoit, The gastrointestinal stability of lipid nanocapsules, ment and characterization of folate anchored saquinavir entrapped PLGA
Int. J. Pharm. 379 (2009) 260265. nanoparticles for anti-tumor activity, Drug Dev. Ind. Pharm. 41 (2015)
[67] S. Peltier, J.-M. Oger, F. Lagarce, W. Couet, J.-P. Benot, Enhanced oral paclitaxel bio- 18881901.
availability after administration of paclitaxel-loaded lipid nanocapsules, Pharm. [97] Y. Cheng, Z. Xu, M. Ma, T. Xu, Dendrimers as drug carriers: applications in different
Res. 23 (2006) 12431250. routes of drug administration, J. Pharm. Sci. 97 (2008) 123143.
[68] P. Sharma, S. Garg, Pure drug and polymer based nanotechnologies for the im- [98] K. Jain, A.K. Verma, P.R. Mishra, N.K. Jain, Characterization and evaluation of
proved solubility, stability, bioavailability and targeting of anti-HIV drugs, Adv. amphotericin B loaded MDP conjugated poly (propylene imine) dendrimers,
Drug Deliv. Rev. 62 (2010) 491502. Nanomedicine 11 (2015) 705713.
[69] H.L. Wong, N. Chattopadhyay, X.Y. Wu, R. Bendayan, Nanotechnology applications [99] H.M. Teow, Z. Zhou, M. Najlah, S.R. Yusof, N.J. Abbott, A. D'Emanuele, Delivery of
for improved delivery of antiretroviral drugs to the brain, Adv. Drug Deliv. Rev. 62 paclitaxel across cellular barriers using a dendrimer-based nanocarrier, Int. J.
(2010) 503517. Pharm. 441 (2013) 701711.
[70] S. Gunaseelan, K. Gunaseelan, M. Deshmukh, X. Zhang, P.J. Sinko, Surface modica- [100] B. Devarakonda, D.P. Otto, A. Judefeind, R.A. Hill, M.M. de Villiers, Effect of pH on
tions of nanocarriers for effective intracellular delivery of anti-HIV drugs, Adv. the solubility and release of furosemide from polyamidoamine (PAMAM) dendri-
Drug Deliv. Rev. 62 (2010) 518531. mer complexes, Int. J. Pharm. 345 (2007) 142153.
[71] E. Roger, F. Lagarce, E. Garcion, J.-P. Benoit, Biopharmaceutical parameters to con- [101] B. Van Eerdenbrugh, G. Van den Mooter, P. Augustijns, Top-down production of
sider in order to alter the fate of nanocarriers after oral delivery, Nanomedicine 5 drug nanocrystals: nanosuspension stabilization, miniaturization and transforma-
(2010) 287306. tion into solid products, Int. J. Pharm. 364 (2008) 6475.
[72] D. Bazile, C. Prud'homme, M.T. Bassoullet, M. Marlard, G. Spenlehauer, M. Veillard, [102] P. Kocbek, S.A. Baumgartner, J. Kristl, Preparation and evaluation of
Stealth Me. PEG-PLA nanoparticles avoid uptake by the mononuclear phagocytes nanosuspensions for enhancing the dissolution of poorly soluble drugs, Int. J.
system, J. Pharm. Sci. 84 (1995) 493498. Pharm. 312 (2006) 179186.
[73] G. Gaucher, P. Satturwar, M.-C. Jones, A. Furtos, J.-C. Leroux, Polymeric micelles for [103] M. Sakai, T. Imai, H. Ohtake, H. Azuma, M. Otagiri, Effects of absorption enhancers
oral drug delivery, Eur. J. Pharm. Biopharm. 76 (2010) 147158. on the transport of model compounds in Caco-2 cell monolayers: assessment by
[74] M.-C. Jones, J.-C. Leroux, Polymeric micellesa new generation of colloidal drug confocal laser scanning microscopy, J. Pharm. Sci. 86 (1997) 779785.
carriers, Eur. J. Pharm. Biopharm. 48 (1999) 101111. [104] A.K. Iyer, G. Khaled, J. Fang, H. Maeda, Exploiting the enhanced permeability and
[75] K. Shao, R. Huang, J. Li, L. Han, L. Ye, J. Lou, C. Jiang, Angiopep-2 modied PE-PEG retention effect for tumor targeting, Drug Discov. Today 11 (2006) 812818.
based polymeric micelles for amphotericin B delivery targeted to the brain, J. Con- [105] H. Zhang, C.P. Hollis, Q. Zhang, T. Li, Preparation and antitumor study of
trol. Release 147 (2010) 118126. camptothecin nanocrystals, Int. J. Pharm. 415 (2011) 293300.
[76] R.D. Dabholkar, R.M. Sawant, D.A. Mongayt, P.V. Devarajan, V.P. Torchilin, [106] A. des Rieux, V. Fievez, I. Thate, J. Mast, V. Prat, Y.-J. Schneider, An improved in
Polyethylene glycolphosphatidylethanolamine conjugate (PEGPE)-based vitro model of human intestinal follicle-associated epithelium to study nanoparti-
mixed micelles: some properties, loading with paclitaxel, and modulation cle transport by M cells, Eur. J. Pharm. Sci. 30 (2007) 380391.
of P-glycoprotein-mediated efux, Int. J. Pharm. 315 (2006) 148157. [107] R.H. Muller, C.M. Keck, Challenges and solutions for the delivery of biotech drugs-a
[77] S.C. Kim, D.W. Kim, Y.H. Shim, J.S. Bang, H.S. Oh, S.W. Kim, M.H. Seo, In vivo evalu- review of drug nanocrystal technology and lipid nanoparticles, J. Biotechnol. 113
ation of polymeric micellar paclitaxel formulation: toxicity and efcacy, J. Control. (2004) 151170.
Release 72 (2001) 191202. [108] L. Gao, G. Liu, J. Ma, X. Wang, L. Zhou, X. Li, Drug nanocrystals: in vivo perfor-
[78] J.Y. Kim, S. Kim, M. Papp, K. Park, R. Pinal, Hydrotropic solubilization of poorly mances, J. Control. Release 160 (2012) 418430.
water-soluble drugs, J. Pharm. Sci. 99 (2010) 39533965. [109] R. Shegokar, R.H. Mller, Nanocrystals: industrially feasible multifunctional formu-
[79] L. Plapied, N. Duhem, A. des Rieux, V. Prat, Fate of polymeric nanocarriers for oral lation technology for poorly soluble actives, Int. J. Pharm. 399 (2010) 129139.
drug delivery, Curr. Opin. Colloid Interface Sci. 16 (2011) 228237. [110] V.K. Pawar, Y. Singh, J.G. Meher, S. Gupta, M.K. Chourasia, Engineered nanocrystal
[80] J. Iqbal, F. Sarti, G. Perera, A. Bernkop-Schnrch, Development and in vivo technology: in-vivo fate, targeting and applications in drug delivery, J. Control. Re-
evaluation of an oral drug delivery system for paclitaxel, Biomaterials 32 lease 183 (2014) 5166.
(2011) 170175. [111] O. Kayser, C. Olbrich, V. Yardley, A.F. Kiderlen, S.L. Croft, Formulation of
[81] M. Ageros, V. Zabaleta, S. Espuelas, M. Campanero, J. Irache, Increased oral bio- amphotericin B as nanosuspension for oral administration, Int. J. Pharm. 254
availability of paclitaxel by its encapsulation through complex formation with cy- (2003) 7375.
clodextrins in poly (anhydride) nanoparticles, J. Control. Release 145 (2010) 28. [112] K. Patel, A. Patil, M. Mehta, V. Gota, P. Vavia, Oral delivery of paclitaxel nanocrystal
[82] V. Zabaleta, G. Ponchel, H. Salman, M. Ageros, C. Vauthier, J.M. Irache, Oral admin- (PNC) with a dual Pgp-CYP3A4 inhibitor: preparation, characterization and antitu-
istration of paclitaxel with pegylated poly (anhydride) nanoparticles: permeability mor activity, Int. J. Pharm. 472 (2014) 214223.
and pharmacokinetic study, Eur. J. Pharm. Biopharm. 81 (2012) 514523. [113] D. Peer, J.M. Karp, S. Hong, O.C. Farokhzad, R. Margalit, R. Langer, Nanocarriers as an
[83] S.-S. Feng, L. Mei, P. Anitha, C.W. Gan, W. Zhou, Poly (lactide)vitamin E derivative/ emerging platform for cancer therapy, Nat. Nanotechnol. 2 (2007) 751760.
montmorillonite nanoparticle formulations for the oral delivery of Docetaxel, Bio- [114] F. Liu, J.-Y. Park, Y. Zhang, C. Conwell, Y. Liu, S.R. Bathula, L. Huang, Targeted cancer
materials 30 (2009) 32973306. therapy with novel high drug-loading nanocrystals, J. Pharm. Sci. 99 (2010)
[84] Z. Zhang, S.-S. Feng, Self-assembled nanoparticles of poly (lactide)vitamin E TPGS 35423551.
copolymers for oral chemotherapy, Int. J. Pharm. 324 (2006) 191198. [115] P. Puligujja, S.S. Balkundi, L.M. Kendrick, H.M. Baldridge, J.R. Hilaire, A.N. Bade, P.K.
[85] H. Chen, Y. Zheng, G. Tian, Y. Tian, X. Zeng, G. Liu, K. Liu, L. Li, Z. Li, L. Mei, Oral de- Dash, G. Zhang, L.Y. Poluektova, S. Gorantla, Pharmacodynamics of long-acting folic
livery of DMAB-modied docetaxel-loaded PLGA-TPGS nanoparticles for cancer acid-receptor targeted ritonavir-boosted atazanavir nanoformulations, Biomate-
chemotherapy, Nanoscale Res. Lett. 6 (2010) 1. rials 41 (2015) 141150.
[86] Y. Dong, S.-S. Feng, Poly (d, l-lactide-co-glycolide)/montmorillonite nanoparticles [116] Y. Liu, L. Huang, F. Liu, Paclitaxel nanocrystals for overcoming multidrug resistance
for oral delivery of anticancer drugs, Biomaterials 26 (2005) 60686076. in cancer, Mol. Pharm. 7 (2010) 863869.
[87] C. Wang, P.C. Ho, L.Y. Lim, Wheat germ agglutinin-conjugated PLGA nanoparticles [117] L. Wang, Z. Liu, D. Liu, C. Liu, Z. Juan, N. Zhang, Docetaxel-loaded-lipid-based-
for enhanced intracellular delivery of paclitaxel to colon cancer cells, Int. J. Pharm. nanosuspensions (DTX-LNS): preparation, pharmacokinetics, tissue distribution
400 (2010) 201210. and antitumor activity, Int. J. Pharm. 413 (2011) 194201.
94 R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195

[118] A. Lemke, A.F. Kiderlen, B. Petri, O. Kayser, Delivery of amphotericin B characterization and transport mechanism across the intestinal barrier, Mol. Pharm.
nanosuspensions to the brain and determination of activity against Balamuthia 7 (2010) 11691182.
mandrillaris amebas, Nanomedicine 6 (2010) 597603. [150] J.H. Schellens, M.M. Malingr, C.M.F. Kruijtzer, H. Bardelmeijer, O. van Tellingen,
[119] Y. Wu, A. Loper, E. Landis, L. Hettrick, L. Novak, K. Lynn, C. Chen, K. Thompson, R. A.H. Schinkel, J.H. Beijnen, Modulation of oral bioavailability of anticancer drugs:
Higgins, U. Batra, The role of biopharmaceutics in the development of a clinical from mouse to man, Eur. J. Pharm. Sci. 12 (2000) 103110.
nanoparticle formulation of MK-0869: a beagle dog model predicts improved bio- [151] J. Van Asperen, O. Van Tellingen, A. Sparreboom, A. Schinkel, P. Borst, W. Nooijen, J.
availability and diminished food effect on absorption in human, Int. J. Pharm. 285 Beijnen, Enhanced oral bioavailability of paclitaxel in mice treated with the P-glyco-
(2004) 135146. protein blocker SDZ PSC 833, Br. J. Cancer 76 (1997) 1181.
[120] A. Karakucuk, N. Celebi, Z.S. Teksin, Preparation of ritonavir nanosuspensions by [152] A. Tsuji, I. Tamai, Carrier-mediated intestinal transport of drugs, Pharm. Res. 13
microuidization using polymeric stabilizers: I. A Design of Experiment approach, (1996) 963977.
Eur. J. Pharm. Sci. (2016). [153] K.-J. Lee, R. Mower, T. Hollenbeck, J. Castelo, N. Johnson, P. Gordon, P.J. Sinko, K.
[121] P. Martin, M. Giardiello, T.O. McDonald, D. Smith, M. Siccardi, S.P. Rannard, A. Holme, Y.-H. Lee, Modulation of nonspecic binding in ultraltration protein bind-
Owen, Augmented inhibition of CYP3A4 in human primary hepatocytes by ritona- ing studies, Pharm. Res. 20 (2003) 10151021.
vir solid drug nanoparticles, Mol. Pharm. 12 (2015) 35563568. [154] M.B. Bolger, I.S. Haworth, A.K. Yeung, D. Ann, H. von Grafenstein, S. Hamm-Alvarez,
[122] S. Jain, J.M. Sharma, A.K. Jain, R.R. Mahajan, Surface-stabilized lopinavir nanoparti- C.T. Okamoto, K.J. Kim, S.K. Basu, S. Wu, Structure, function, and molecular modeling
cles enhance oral bioavailability without coadministration of ritonavir, approaches to the study of the intestinal dipeptide transporter PepT1, J. Pharm. Sci. 87
Nanomedicine 8 (2013) 16391655. (1998) 12861291.
[123] T.-k. Wu, S.-Y. Lin, H.-L. Lin, Y.-T. Huang, Simultaneous DSC-FTIR [155] A. Rice, M.L. Michaelis, G. Georg, Y. Liu, B. Turunen, K.L. Audus, Overcoming the
microspectroscopy used to screen and detect the co-crystal formation in real blood-brain barrier to taxane delivery for neurodegenerative diseases and brain
time, Bioorg. Med. Chem. Lett. 21 (2011) 31483151. tumors, J. Mol. Neurosci. 20 (2003) 339343.
[124] N.R. Goud, S. Gangavaram, K. Suresh, S. Pal, S.G. Manjunatha, S. Nambiar, A. Nangia, [156] B.S. Anand, S. Katragadda, A.K. Mitra, Pharmacokinetics of novel dipeptide ester
Novel furosemide cocrystals and selection of high solubility drug forms, J. Pharm. prodrugs of acyclovir after oral administration: intestinal absorption and liver me-
Sci. 101 (2012) 664680. tabolism, J. Pharmacol. Exp. Ther. 311 (2004) 659667.
[125] S. Spireas, S. Sadu, Enhancement of prednisolone dissolution properties using [157] C. Rousselle, P. Clair, J.-M. Lefauconnier, M. Kaczorek, J.-M. Scherrmann, J. Temsamani,
liquisolid compacts, Int. J. Pharm. 166 (1998) 177188. New advances in the transport of doxorubicin through the blood-brain barrier by a
[126] S. Spireas, S. Sadu, R. Grover, In vitro release evaluation of hydrocortisone liquisolid peptide vector-mediated strategy, Mol. Pharmacol. 57 (2000) 679686.
tablets, J. Pharm. Sci. 87 (1998) 867872. [158] R. Jain, S. Agarwal, S. Majumdar, X. Zhu, D. Pal, A.K. Mitra, Evasion of P-gp mediated
[127] Y. Javadzadeh, M. Siahi-Shadbad, M. Barzegar-Jalali, A. Nokhodchi, Enhancement of cellular efux and permeability enhancement of HIV-protease inhibitor saquinavir
dissolution rate of piroxicam using liquisolid compacts, Il Farmaco 60 (2005) by prodrug modication, Int. J. Pharm. 303 (2005) 819.
361365. [159] R. Jain, S. Duvvuri, V. Kansara, N.K. Mandava, A.K. Mitra, Intestinal absorption of
[128] V. Yadav, A. Yadav, Improvement of solubility and dissolution of indomethacin by novel-dipeptide prodrugs of saquinavir in rats, Int. J. Pharm. 336 (2007) 233240.
liquisolid and compaction granulation technique, Drugs 5 (2009) 8. [160] R. Jain, S. Agarwal, N.K. Mandava, Y. Sheng, A.K. Mitra, Interaction of dipeptide
[129] V. Yadav, A. Yadav, Liquisolid granulation technique for tablet manufacturing: an prodrugs of saquinavir with multidrug resistance protein-2 (MRP-2): evasion of
overview, J. Pharm. Res. 2 (2009). MRP-2 mediated efux, Int. J. Pharm. 362 (2008) 4451.
[130] V.B. Yadav, A.V. Yadav, Enhancement of solubility and dissolution rate of BCS class [161] S. Agarwal, S. Boddu, R. Jain, S. Samanta, D. Pal, A.K. Mitra, Peptide prodrugs: im-
II pharmaceuticals by nonaqueous granulation technique, Int. J. Pharm. Res. Dev. 1 proved oral absorption of lopinavir, a HIV protease inhibitor, Int. J. Pharm. 359
(2010) 112. (2008) 714.
[131] Y. Javadzadeh, M.R. Siahi, S. Asnaashari, A. Nokhodchi, An investigation of physico- [162] M. Rouquayrol, B. Gaucher, D. Roche, J. Greiner, P. Vierling, Transepithelial trans-
chemical properties of piroxicam liquisolid compacts, Pharm. Dev. Technol. 12 port of prodrugs of the HIV protease inhibitors saquinavir, indinavir, and nelnavir
(2007) 337343. across Caco-2 cell monolayers, Pharm. Res. 19 (2002) 17041712.
[132] R.H. Fahmy, M.A. Kassem, Enhancement of famotidine dissolution rate through [163] S.A. Galindo-Rodrguez, F. Puel, S. Brianon, E. Allmann, E. Doelker, H. Fessi, Com-
liquisolid tablets formulation: in vitro and in vivo evaluation, Eur. J. Pharm. parative scale-up of three methods for producing ibuprofen-loaded nanoparticles,
Biopharm. 69 (2008) 9931003. Eur. J. Pharm. Sci. 25 (2005) 357367.
[133] B. Akinlade, A.A. Elkordy, E.A. Essa, S. Elhagar, Liquisolid systems to improve the [164] D.L. Marchisio, L. Rivautella, A.A. Barresi, Design and scale-up of chemical reactors
dissolution of furosemide, Sci. Pharm. 78 (2010) 325. for nanoparticle precipitation, AICHE J. 52 (2006) 18771887.
[134] S. Burra, S.K. Galipelly, Enhancement of solubility and dissolution rate of frusemide [165] A. Basu, M. Nazarkovsky, R. Ghadi, W. Khan, A.J. Domb, Poly (lactic acid)-based
through liquisolid technique, Pharm. Lett. 2 (2010) 321328. nanocomposites, Polym. Adv. Technol. (2016).
[135] X. Zhao, Y. Zhou, S. Potharaju, H. Lou, H. Sun, E. Brunson, H. Almoazen, J. Johnson, De- [166] A.S. Rathore, H. Winkle, Quality by design for biopharmaceuticals, Nat. Biotechnol.
velopment of a self micro-emulsifying tablet of cyclosporine-a by the liquisolid com- 27 (2009) 26.
pact technique, Int. J. Pharm. Sci. Res. 2 (2011) 2299. [167] D.P. Petrides, A. Koulouris, P.T. Lagonikos, The role of process simulation in phar-
[136] K.A. Khaled, Y.A. Asiri, Y.M. El-Sayed, In vivo evaluation of hydrochlorothiazide maceutical process development and product commercialization, Pharm. Eng. 22
liquisolid tablets in beagle dogs, Int. J. Pharm. 222 (2001) 16. (2002) 5665.
[137] A. Serajuddin, P.C. Sheen, D. Mufson, D.F. Bernstein, M.A. Augustine, Effect of vehi- [168] R.H. Myers, D.C. Montgomery, C.M. Anderson-Cook, Response Surface Methodolo-
cle amphiphilicity on the dissolution and bioavailability of a poorly water-soluble gy: Process and Product Optimization Using Designed Experiments, John Wiley &
drug from solid dispersions, J. Pharm. Sci. 77 (1988) 414417. Sons, 2016.
[138] S.R. Vippagunta, K.A. Maul, S. Tallavajhala, D.J. Grant, Solid-state characterization of [169] S.C. Porter, R.P. Verseput, C.R. Cunningham, Process optimization using design of
nifedipine solid dispersions, Int. J. Pharm. 236 (2002) 111123. experiments, Pharm. Technol. 21 (1997) 6071.
[139] Y. Chen, G. Zhang, J. Neilly, K. Marsh, D. Mawhinney, Y. Sanzgiri, Enhancing the bio- [170] N. Desai, Challenges in development of nanoparticle-based therapeutics, AAPS J. 14
availability of ABT-963 using solid dispersion containing Pluronic F-68, Int. J. (2012) 282295.
Pharm. 286 (2004) 6980. [171] C. Vauthier, K. Bouchemal, Methods for the preparation and manufacture of poly-
[140] S.X. Yin, M. Franchini, J. Chen, A. Hsieh, S. Jen, T. Lee, M. Hussain, R. Smith, Bioavail- meric nanoparticles, Pharm. Res. 26 (2009) 10251058.
ability enhancement of a COX-2 inhibitor, BMS-347070, from a nanocrystalline dis- [172] H.R. Lakkireddy, D. Bazile, Building the design, translation and development prin-
persion prepared by spray-drying, J. Pharm. Sci. 94 (2005) 15981607. ciples of polymeric nanomedicines using the case of clinically advanced poly
[141] E.T. Cole, Liquid-lled hard gelatin capsules, Pharm. Technol. Int. 1 (1989) 2933. (lactide (glycolide))poly (ethylene glycol) nanotechnology as a model: an indus-
[142] J. Boudreaux, D. Hayes, S. Mizrahi, P. Maggiore, J. Blazek, D. Dick, Use of Water-Sol- trial viewpoint, Adv. Drug Deliv. Rev. (2016).
uble Liquid Vitamin E to Enhance Cyclosporine Absorption in Children after Liver [173] M.A. Vetten, C.S. Yah, T. Singh, M. Gulumian, Challenges facing sterilization and
Transplant, Transplantation Proceedings 1993, p. 1875. depyrogenation of nanoparticles: effects on structural stability and biomedical ap-
[143] R.J. Sokol, N. Butler-Simon, C. Conner, J.E. Heubi, F.R. Sinatra, F.J. Suchy, M.B. Heyman, J. plications, Nanomedicine 10 (2014) 13911399.
Perrault, R.J. Rothbaum, J. Levy, Multicenter trial of d-alpha-tocopheryl polyethylene [174] R. Aitken, M. Chaudhry, A. Boxall, M. Hull, Manufacture and use of nanomaterials:
glycol 1000 succinate for treatment of vitamin E deciency in children with chronic current status in the UK and global trends, Occup. Med. 56 (2006) 300306.
cholestasis, Gastroenterology 104 (1993) 17271735. [175] J.I. Hare, T. Lammers, M.B. Ashford, S. Puri, G. Storm, S.T. Barry, Challenges and
[144] T. Chang, L.Z. Benet, M.F. Hebert, The effect of water-soluble vitamin E on cyclosporine strategies in anti-cancer nanomedicine development: an industry perspective,
pharmacokinetics in healthy volunteers, Clin. Pharm. Ther. 59 (1996) 297303. Adv. Drug Deliv. Rev. (2016).
[145] S. Sinha, M. Ali, S. Baboota, A. Ahuja, A. Kumar, J. Ali, Solid dispersion as an ap- [176] H. Ragelle, F. Danhier, V. Prat, R. Langer, D.G. Anderson, Nanoparticle-based drug
proach for bioavailability enhancement of poorly water-soluble drug ritonavir, delivery systems: a commercial and regulatory outlook as the eld matures, Expert
AAPS PharmSciTech 11 (2010) 518527. Opin. Drug Deliv. (2016) 114.
[146] M.A. Ibrahim, M. El-Badry, Formulation of immediate release pellets containing [177] P. Maincent, The regulatory environment: the challenges for lipid-based formula-
famotidine solid dispersions, Saudi Pharm. J. 22 (2014) 149156. tion, Bull. Tech. Gattefoss 100 (2007) 4749.
[147] V. Risovic, M. Boyd, E. Choo, K. Wasan, Effect of various lipid-based oral formula- [178] M.-L. Chen, Lipid excipients and delivery systems for pharmaceutical develop-
tions on plasma and tissue concentrations and renal toxicity of amphotericin B ment: a regulatory perspective, Adv. Drug Deliv. Rev. 60 (2008) 768777.
within male rats, Antimicrob. Agents Chemother. 47 (2003) 33393342. [179] H. Shrestha, R. Bala, S. Arora, Lipid-based drug delivery systems, J. Pharm. 2014
[148] V. Risovic, K. Sachs-Barrable, M. Boyd, K.M. Wasan, Potential mechanisms by which (2014).
Peceol increases the gastrointestinal absorption of amphotericin B, Drug Dev. Ind. [180] R. Ghadi, A. Ghuge, S. Ghumre, N. Waghmare, V.J. Kadam, Co-crystals: emerging
Pharm. 30 (2004) 767774. approach in pharmaceutical design, Am. J. Pharm. Res. 4 (2014).
[149] Y. Zhang, X. Li, Y. Zhou, Y. Fan, X. Wang, Y. Huang, Y. Liu, Cyclosporin A-loaded poly [181] A.V. Trask, An overview of pharmaceutical cocrystals as intellectual property, Mol.
(ethylene glycol)-b-poly (d, l-lactic acid) micelles: preparation, in vitro and in vivo Pharm. 4 (2007) 301309.
R. Ghadi, N. Dand / Journal of Controlled Release 248 (2017) 7195 95

[182] U. Food, D. Administration, Guidance for Industry: Regulatory Classication of [185] U. Food, D. Administration, Draft Guidance for Industry: Considering whether an
Pharmaceutical Co-crystals, Center for Drug Evaluation and Research, Silver Spring, FDA-regulated Product Involves the Application of Nanotechnology, US FDA, 2011.
US, 2013. [186] S. Tinkle, S.E. McNeil, S. Mhlebach, R. Bawa, G. Borchard, Y.C. Barenholz, L.
[183] M. Joint, EMA reection paper on the development of block copolymer micelle me- Tamarkin, N. Desai, Nanomedicines: addressing the scientic and regulatory gap,
dicinal products, draft, Eur. Union Eur. Med. Agency 44 (2013) 118. Ann. N. Y. Acad. Sci. 1313 (2014) 3556.
[184] V. Sainz, J. Conniot, A.I. Matos, C. Peres, E. Zupani, L. Moura, L.C. Silva, H.F. [187] N. Sadrieh, Overview of CDER Experience with Nanotechnology-related Drugs,
Florindo, R.S. Gaspar, Regulatory aspects on nanomedicines, Biochem. Biophys. Slides for the August, 9, 2012.
Res. Commun. 468 (2015) 504510.

You might also like