Sie sind auf Seite 1von 46

Accepted Manuscript

Title: Lipid-based formulations for oral administration of


poorly water-soluble drugs
Author: Huiling Mu Rene Holm Anette Mullertz
PII:
DOI:
Reference:

S0378-5173(13)00284-6
http://dx.doi.org/doi:10.1016/j.ijpharm.2013.03.054
IJP 13234

To appear in:

International Journal of Pharmaceutics

Received date:
Revised date:
Accepted date:

27-1-2013
28-3-2013
28-3-2013

Please cite this article as: Mu, H., Holm, R., Mullertz, A., Lipid-based formulations
for oral administration of poorly water-soluble drugs, International Journal of
Pharmaceutics (2013), http://dx.doi.org/10.1016/j.ijpharm.2013.03.054
This is a PDF file of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its final form. Please note that during the production process
errors may be discovered which could affect the content, and all legal disclaimers that
apply to the journal pertain.

1
Lipid-based formulations for oral administration of poorly water-soluble drugs

ip
t

Huiling Mu a*, Ren Holma,b, and Anette Mllertza,c

Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen,

Biologics and Pharmaceutical Science, H.Lundbeck A/S, Ottiliavej 9, DK-2500 Valby,

us

Denmark.

10

Bioneer:FARMA, Danish Drug Development Center, Department of Pharmacy, Faculty of

an

cr

Universitetsparken 2, DK-2100 Copenhagen, Denmark.

Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100

Ac
ce
p

15

te

Copenhagen, Denmark.

*Correspondence: Dr. H. Mu, Department of Pharmacy, Faculty of Health and Medical Sciences,
20

University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark. E-mail:


huiling.mu@sund.ku.dk

Page 1 of 45

2
Abstract
Lipid-based drug delivery systems have shown great potentials in oral delivery of poorly water25

soluble drugs, primarily for lipophilic drugs, with several successfully marketed products. Pre-

ip
t

dissolving drugs in lipids, surfactants, or mixtures of lipids and surfactants omits the

dissolving/dissolution step, which is a potential rate limiting factor for oral absorption of poorly

cr

water-soluble drugs. Lipids not only vary in structures and physiochemical properties, but also in

30

us

their digestibility and absorption pathway; therefore selection of lipid excipients and dosage form
has a pronounced effect on the biopharmaceutical aspects of drug absorption and distribution

an

both in vitro and in vivo. The aim of this review is to provide an overview of the different lipidbased dosage forms from a biopharmaceutical point of view and to describe effects of lipid

te

Key words: Lipids, lipid-based drug delivery systems, lipid digestion, lymphatic transport, in
vitro lipolysis, poorly water-soluble drug

Ac
ce
p

35

dosage forms and lipid excipients on drug solubility, absorption and distribution.

Page 2 of 45

3
1. Introduction
Oral drug administration is desirable due to good patient convenience and consequently better
compliance. To be absorbed from the gastrointestinal (GI) tract, a drug needs to be dissolved in
the GI fluids; this is a problem for the increasing number of poorly water-soluble drug candidates

ip
t

40

that are in development in the pharmaceutical industry. However, it seems that a high

cr

permeability is maintained for most of these compounds, rendering them class II drugs in the

us

Biopharmaceutics Classification System (BCS) (Amidon et al., 1995; Lipinski 2000; Lipinski et

45

limiting step for the absorption of these drugs.

an

al., 1997; Yu et al., 2002). Thus the solubility and/or dissolution rate in the GI tract often is the

The interests on lipid-based drug delivery systems (LBDDS) have increased over the past two

decades as a function of identification of these pharmaceutically difficult candidates, and

50

te

increased even further after successful launch of lipid-based oral pharmaceutical products,
including in particular cyclosporine A, marketed as SandimmuneTM and NeoralTM. One of the

Ac
ce
p

advantages of LBDDS is that drug molecules are pre-dissolved in lipid excipients, avoiding a
potentially rate limiting dissolution step in the GI tract, thereby achieving an increased and
consistent bioavailability (Chakraborty et al., 2009; Drewe et al., 1992; Gershanik and Benita
2000; Hauss D.J. 2007; Kohli et al., 2010; Porter et al., 2008; Strickley 2004).
55

To develop LBDDS, several complex biological processes have to be taken into account, such as
digestion of lipid excipients, formation of different colloid phases during lipid digestion, and
transfer of the drug between these colloid phases. Several reviews of lipid-based formulations are
available, each focusing on different aspects of lipids in drug delivery (Chakraborty et al.,2009;

Page 3 of 45

4
60

Fricker et al., 2010; Gursoy and Benita 2004; Hauss D.J.2007; Kuentz 2011; Kohli et al.,2010;
Mllertz et al., 2010; Porter et al., 2007; Pouton 2000; Pouton 2006; Porter et al.,2008; Rahman
et al., 2011; Shukla et al., 2011; Singh et al., 2009a; Singh et al., 2011; Yanez et al., 2011).

ip
t

Pouton (2006, 2000) proposed a Lipid Formulations Classification System (LFCS) and

categorized lipid-based formulations into four different types according to their compositions.
Porter and colleagues summarised lipid delivery systems with focus on self-emulsifying delivery

cr

65

us

system (SEDDS) and assessment of lipid-based formulations using in vitro lipolysis (Porter et
al.,2008), and provided a good overview on lipid digestion and drug solubilisation in the small

an

intestine as well as lymphatic transport (Porter et al.,2007). Lipid excipients have been reviewed
by Hauss in context of their applications in lipid-based formulations (Hauss D.J.2007) and
application of phospholipids in oral drug delivery has been reviewed by Fricker et al. (2010).

70

Singh et al. have made a general review covering SEDDS and solid dispersions (Singh et

te

al.,2011). In a recent review a rational strategy for the development of lipid and surfactant based
drug delivery system was suggested (Mllertz et al.,2010). In connection to this the LFCS

75

Ac
ce
p

proposed by Pouton was evaluated and considerations suggested in particular reflecting the
differences between triglycerides (TG) and partial glycerides.

This review presents the recent progress on liquid lipid-based dosage forms, with emphasis on
the formulation forms from a biopharmaceutical point of view. This includes simple
considerations on drug selection and lipid digestion related with lipid structure, and an overview
80

of different lipid-based dosage forms, from simple lipid solutions to advanced SEDDS and
solidifying lipid formulations.

Page 4 of 45

5
2. Selection of compounds for lipid based formulations
Compounds with a low aqueous solubility, i.e. belonging to the BCS class II and IV, are
85

frequently discussed in relation to LBDDS. Compounds may fall into BCS II and IV for different

ip
t

physical chemical reasons; therefore it can be helpful to identify the etiology of the poor

solubility. A number of poorly water-soluble compounds are regarded as brick dust and cannot

cr

be formulated as LBDDS because of their tight crystal lattice; another group of compounds

90

us

possesses high lipophilicity (logP) and much lower melting points, the so-called grease-balls.
Obviously, a continuum exists between these two simplified classes of poorly soluble

an

compounds, with most drug molecules not fitting either extreme. If the molecule has the
characteristics of a grease-ball and traditional formulation approaches do not provide adequate

bioavailability, solubility enhancement through the use of surfactant and/or lipid based excipients

lipids, but may have considerable higher solubility in surfactants and co-solvents. Therefore not

te

95

may be useful. If the compound is a brick-dust molecule, it typically has a low solubility in

all compounds with poor aqueous solubility and/or a high log P will have a good solubility in

Ac
ce
p

excipients that are suitable for LBDDS.

Mllertz and co-workers (2010) recently summarised a number of commercial formulations. Of


100

the 25 formulations classified 13 were LFCS class I, three were class III, and nine were class IV.
A few simple descriptors for the compounds and their melting points are presented in Table 1,
demonstrating that the simple classification into grease-balls and brick-dust is not always
predictive in the selection of the type of lipid-based formulations. Rane and Anderson (2008)
recently provided a review of the different theoretical models developed to predict the solubility

105

of compounds in lipids or lipid mixtures. At present, no model is capable of predicting the

Page 5 of 45

6
solubility of a drug molecule in lipids, but if new theories can be developed that takes the
specific and non-specific interactions between the compounds and the lipid excipients into the

110

ip
t

consideration, they may hold potential for future predictions.

When defining the formulation space for LBDDS the use of Design of Experiments (DoE) is a

cr

good and systematic approach. Output parameters often include drug solubility, colloid

us

structures of the aqueous dispersions, and in the case of SEDDS droplet size of the formed
emulsion. When incorporating the solubility as an output in DoE, higher solubility may be

115

an

achieved in the formulation mixtures than in individual excipients (Holm et al., 2006a), even
though drug solubility in lipid mixtures can be estimated as a simple weighted average of the

drug solubility in individual ingredients (Sacchetti and Nejati 2012). If the solubility of a

compound is low, the solubility should generally be screened. Therefore preformulation

te

measurements of solubility in excipients and mixtures hereof are the most useful parameters to

120

Ac
ce
p

determine if lipid-based formulation is a feasible strategy for a given compound.

3. Physicochemical properties of lipids


Lipids are generally defined as compounds containing fatty acids and can be separated into
different classes (Cast and Hamilton 1999; Castera 1995; Mu 2005). Small (1968) has classified
lipids based on their interactions in aqueous systems and their behaviour in the waterair
125

interface. Free fatty acids (FFA), diglycerides (DG), and TG are classified as class I polar lipids
because they possess some surface solubility and can form stable monolayers on the surface
(Small1968). Monoglycerides (MG) and phospholipids are classified as class II polar lipids,
which can form well-defined liquid crystalline phases in the bulk (Small1968). Shimada and

Page 6 of 45

7
Ohashi (2003) studied the interfacial and emulsifying properties of DG and MG; they found that
130

addition of DG to TG reduced the interfacial tension, and addition of MG to TG had a more


profound effect in the reduction of the interfacial tension. Pitzalis et al. (2000) characterised the

ip
t

ternary phase diagrams of MG and DG in water and found that MG formed a lamellar phase and
two types of bi-continuous cubic phases, which further confirms that MG is different from TG in

classified as neutral lipids in food science and lipid chemistry (Christie 1985; Heinz 2012), their

us

135

cr

the formation of microstructures in aqueous media. Even though MG, DG, and TG are normally

polarity is increasing in the order of TG, DG, and MG (Christie1985; Mu et al., 2000a).

an

Therefore TG, DG and MG are different not only in their structures, but also different in their

In addition to the differences of their physiochemical properties, the in vivo digestion processes

140

surface activities and polarities.

te

of TG, DG and MG are also different. Digestion of lipids already starts in the stomach; pre-

Ac
ce
p

duodenal lipases partially hydrolyse TG to 1,2(2,3)-DG and FFA, with up to 30% of TG being
digested in the stomach, depending on the fatty acid composition of the TG (Armand 2007;
Hamosh 1979; Hamosh and Scow 1973; Smith et al., 1986). 1,2(2,3)-DG are further digested in
145

the small intestine by other lipases such as pancreatic lipase to 2-MG and FFA (Christophe 2004;
Mu and Hy 2004; Tso 1985). The intramolecular structure of TG affects lipid digestion and
absorption (Mu and Porsgaard 2005), TG and DG are also digested and absorbed differently.
Watanabe and Tokimitsu (2004) investigated the digestion and absorption of TG and DG in rats
by duodenal infusion of 14C-labeled lipids. Ten min after infusion of TG and 1,3-DG, the

150

lipolytic products of TG contained 10% of 1,2(2,3)-DG, 28% of 2-MG and 37% of FFA,
whereas the lipolytic products of 1,3-DG contained 26% of 1(3)-MG and 51 % of FFA. The high

Page 7 of 45

8
ratio of FFA/1(3)-MG in the lumen infused with DG suggested that substantial amounts of 1(3)MG were further converted to FFA and glycerol (Watanabe et al.,2004). A study in cannulated
pigs also showed increased portal transport of glycerol after intake of DG oil, supporting the
further hydrolysis of 1(3)-MG (Kristensen et al., 2006). Since 1(3)-MG is often used as lipid

ip
t

155

excipients alone or as mixtures with DG and TG, the difference of lipid digestion in the GI tract

cr

will affect the solubility of drug molecules in the GI fluids. Understanding lipid structures and

4. Lipid-based drug delivery systems

an

160

us

digestibility is therefore an important part of formulation considerations for LBDDS.

LBDDS encompass a range of formulations, from simple lipid solutions to advanced SEDDS,

which are presented separately below. Selection of lipid excipients does not only affect drug

solubility in the formulations, but also the solubilisation of drugs in the GI tract during lipid

165

te

digestion, as well as the absorption and bioavailability of drugs. Figure 1 is a simplified


illustration of digestion of lipid-based formulations and formation of micelles in the GI tract.

Ac
ce
p

Partially digested lipids from the stomach are solubilised in the GI fluids in the presence of both
bile and pancreatic juice, fine emulsions are further converted to bile salt micelles and lamellar
vesicles (Mllertz et al., 2012; Tso 1994). These particles deliver digested lipids and drug
molecules to enterocytes, and both the lipid and drug molecules are subsequently absorbed. The
170

dispersion of lipids in the GI fluids affects the kinetics of lipid digestion and absorption
(Armand2007; Singh et al., 2009b), as well as solubility and absorption of drugs from lipidbased formulations (Kaukonen et al., 2004a; Kaukonen et al., 2004b).

Page 8 of 45

9
In vitro lipolysis is often used for the evaluation of LBDDS and the impact of lipid digestion on
175

drug solubilisation, as well as characterisation of lipolysis products formed (Fatouros et al., 2007;
Kleberg et al., 2010a; Larsen et al., 2011; Larsen et al., 2008; Porter et al.,2008; Sassene et al.,

ip
t

2010; Thomas et al., 2012c; Zangenberg et al., 2001a; Zangenberg et al., 2001b). Different

biorelevant media have been used for in vitro lipolysis (Kleberg et al., 2010b). The lipolysis is

and the lipid-based formulation, lipid hydrolysis is initiated by addition of pancreatic lipases

us

180

cr

typically carried out in a thermo-controlled reaction vessel containing simulated intestinal fluids

(Figure 2A). Lipolysis samples can be separated by ultracentrifugation into at least two phases,

an

liquid- and pellet-phase; the pellet-phase can contain both calcium soaps of fatty acids and
precipitated drugs (Figure 2B). And the composition of excipients can affect drug distribution

absorption and more discussions about drug precipitation from lipid-based formulations can be

185

between the different phases (Figure 2C) (Ren et al., 2012). Drug precipitation may affect its

te

found later in this section. More details about the in vitro lipolysis model and its application in

Ac
ce
p

the characterisation of LBDDS can be found in a recent review by Thomas et al. (2012b).

In the case of targeting drugs to intestinal lymphatic transport, long-chain fatty acids will be a
190

better choice because they can stimulate formation and secretion of chylomicrons and improve
lymphatic transport of lipids (Holm et al., 2006b; Kalogeris et al., 1996; Mu et al.,2005; Mu and
Hoy 2001) as well as lipophilic compounds (Caliph et al., 2000; Charman and Stella 1986a;
Charman and Stella 1986b; Gershkovich et al., 2007; Porter et al.,2007). The chain-length of
fatty acids affects the distribution of lipids between the lymphatics and the portal vein (Mu and

195

Hy 2000b). Figure 3 shows the similar tendency for the increased lymphatic transport of lipids
and halofantrine solubilised in lipids with the increasing chain-length of fatty acids, illustrating

Page 9 of 45

10
that the selection of lipid excipients is one of the key factors for lymphatic transport. Holm et al.
(2003) also demonstrated the importance of intramolecular structure of TG for lymphatic
transport of poorly water-soluble drugs. They compared the lymphatic transport of halofantrine
in cannulated canines after administration SEDDS containing either TG8:0/18:2/8:0 or

ip
t

200

TG18:2/8:0/18:2 and found that the extent of lymphatic transport via the thoracic duct was 18%

cr

and 27% of the dose for TG8:0/18:2/8:0 and TG18:2/8:0/18:2, respectively (Holm et al.,2003).

us

Gershkovich et al (2007) investigated the role of lymphatic transport in the oral bioavailability of
two structurally similar lipophilic drugs with similar logP values, 8.2 and 8.5. The solubility of
Dexanabinol and PRS-211,220 in long-chain TG was 7.9 and 95.8 mg/g, respectively. The

an

205

intestinal lymphatic transport of dexanabinol and PRS-211,220 was 7.5 and 60.7% of the

absorbed dose, respectively (Gershkovich et al.,2007). This is in agreement with a previous study

of lymphatic transport of hexachlorobenzene and 1,1-bis(p-chlorophenyl)-2,2,2-trichloroethane

210

te

(Charman et al.,1986b). These studies demonstrate that drug solubility in lipids is one of the
determining factors for lymphatic drug transport. Lymphatic transport and metabolic enzymes as

Ac
ce
p

well as transporters involved in intestinal lymphatic transport have recently been


comprehensively reviewed by Yanez et al. (2011).

4.1 Lipid solutions


215

Lipid solutions are the simplest lipid-based formulation and several products are already on the
market (Mllertz et al.,2010). Lipid excipients, used in this type of formulations, vary in their
physicochemical properties and their abilities to be digested. The solubility of the drug molecules
in the selected lipid excipients is a critical factor for the success of the formulation. The lipids are
either based upon long-chain fatty acids or medium-chain fatty acids, for instance medium-chain

Page 10 of 45

11
220

TG (MCT) has been used to solubilise efavirenz (SustivaTM), whereas long-chain TG (LCT) such
as sesame oil has been used in Marinol (dronabinol) and One-Alpha (alfacalcidol).

ip
t

Dissolving drugs in lipids can significantly improve the bioavailability of poorly water-soluble
drugs. Seocalcitol is a lipophilic drug with a log P value 4.8. The solubility of seocalcitol in

water is only 20 ng/g, whereas its solubility in MCT and LCT is 5.3 and 1.7 mg/g, respectively

cr

225

us

(Grove et al., 2005). Grove et al. (2005) investigated the bioavailability of seocalcitol in rats
after oral administration its lipid solutions and reported a twofold increase of the bioavailability

an

when seocalcitol was dissolved in lipids compared with a reference formulation containing
propylene glycol, whereas no difference was observed for the MCT and the LCT solutions.
Similar results have been reported for other lipophilic drugs. For instance lipid solutions of

230

anethol trithione (log P=3.8) improved the oral absorption of anethol trithione in rats, but there

235

Ac
ce
p

(Han et al., 2009).

te

were no significant differences between lipid solutions of short-chain TG (SCT), MCT, and LCT

Christensen et al. (2004) studied the in vitro solubility of poorly water-soluble drugs during
lipolysis of their lipid solutions containing either MCT or LCT and found different effects of
fatty acids on solubilisation of drugs in the aqueous phase. No difference was observed for
solubilisation of probucol when it was formulated as MCT or LCT solutions, whereas another
drug (LU 28-179) was solubilised better when it was formulated as LCT solution, suggesting that

240

the properties of drugs can affect their distribution between the different phases during lipolysis
(Christensen et al.,2004). Dahan and Hoffman (2007) also observed that the properties of drugs
can affect their solubilisation during in vitro lipolysis as well as the bioavailability. Lipophilic

Page 11 of 45

12
drug dexamethasone and griseofulvin were dissolved in SCT, MCT or LCT; the in vitro
performance of the different formulations for dexamethasone was similar, whereas a
245

performance rank order of MCT > LCT > SCT was observed for griseofulvin. The in vivo

ip
t

bioavailability of both drugs correlated well with the in vitro data, i.e., LCT = MCT = SCT for
dexamethasone and MCT > LCT > SCT for griseofulvin (Dahan et al.,2007). Recently lipid

cr

solutions of fenofibrate were subjected to in vitro digestion aiming at predicting the fate of the

250

us

drug in the GI tract (Mohsin 2012). MCT was almost completely hydrolysed after 30 min,
however, only 5-7% of fenofibrate were carried in the aqueous phase; whereas 21-36% of

an

fenofibrate were solubilised in the aqueous phase after digestion of the formulation containing

soybean oil.

of oral lipid solutions of halofantrine in beagle dogs and found that in vitro drug solubilisation

te

255

Porter et al. (2004) tried to use the in vitro lipid digestion data to explain the in vivo performance

profiles were markedly dependent on the mass of lipid employed in lipid digestion experiments.

Ac
ce
p

After 60 min in vitro lipolysis, more halofantrine from the MCT formulations with high lipid
loads (ca. 25 mg TG/mL digestion medium) were solubilised in the aqueous medium, whereas at
low lipid concentrations (ca. 5mg/mL digestion medium), LCT formulations provided improved
260

solubilisation capacity. The in vitro digestion and solubilisation data at lower lipid masses were
consistent with the in vivo date where the bioavailability of halofantrine after oral administration
of LCT solution was higher than MCT solutions (Porter et al.,2004). Therefore, when comparing
MCT and LCT containing LBDDS, it seems possible to assess the rank order of in vivo
performance of lipid solutions by the amount of drug solubilised during in vitro lipolysis.

265

Page 12 of 45

13
All in all, lipid solutions can be used to improve the bioavailability of lipophilic drugs. The in
vitro drug solubilisation profiles can be affected by many factors, including lipid excipients, the

270

ip
t

properties of drugs, and the mass of lipid employed in lipid digestion experiments.

4.2. Lipid suspensions

cr

As discussed above, a poor solubility in aqueous media is not necessarily equal to a good

us

solubility in pharmaceutically relevant lipids; hence it is not always possible to dissolve the
needed drug dose in the excipients - even after thorough excipient screening. As an alternative, a

limited attention.

275

an

suspension of the drug in lipids can be used; a formulation approach that has only received

Carrigan and Bates demonstrated for more than 35 years ago that griseofulvin suspended in corn

te

oil resulted in a significantly higher bioavailability compared to administration in an aqueous


suspension, when dosed orally to rats (Carrigan and Bates 1973). In support of this, a clear trend
towards a higher bioavailability of phenytoin was seen when administered in a corn oil

Ac
ce
p

280

suspension compared to an aqueous suspension, though an emulsion performed even better


(Chakrabarti and Belpaire 1978). Abrams et al. (1978) reported a higher absorption of a
lipophilic steroid from a sesame oil solution when compared to an aqueous suspension, but
similar to suspensions in sesame oil. Hargrove et al. (1989) evaluated the influence of particle
285

size and vehicle on the absorption of propesterone in humans. Micronisation of the compound
increased the absorption of the compound significantly, but a suspension of the micronized
compound in oil increased it even further. In a more recent study, Larsen and co-workers
investigated the bioavailability of danazol solubilised or suspended in the PEG based surfactant

Page 13 of 45

14
Labrafil M2125CS (Larsen et al.,2008). The authors reported that a suspension of danazol led
290

to the same oral bioavailability as a lipid solution, from which it was concluded that lipid

ip
t

suspension vehicles may perform just as well as lipid solutions for some compounds.

Altogether this demonstrates that the physical form is important for the absorption, also from a

the situations where the solubility in lipids is insufficient. From an industrial point of view,

us

295

cr

lipid suspension, additionally that this relative unexplored formulation type may be an option in

however, lipid based suspensions are slightly more difficult to manufacture than the solutions, as

an

there is a need for continuous focus on sedimentation and physical stabilization of the suspension.
Alternatively a small capsule containing the drug molecule plus disintegrants can be included in

a capsule containing the lipid, i.e. capsule in a capsule.

300

te

4.3. Self-Emulsifying Drug Delivery Systems

SEDDS and Self-NanoEmulsifying Drug Delivery Systems (SNEDDS) are type II and type III

Ac
ce
p

formulations in the LFCS. Drug molecules are dissolved in SEDDS/SNEDDS preconcentrates in


these formulations. Upon dispersion of the preconcentrates in the GI fluids an emulsion is
305

formed, in the case of SNEDDS, this is a nanoemulsion. Emulsions and nanoemulsions often
contribute to an uniform and reproducible oral bioavailability of the drug in both the fasted and
fed state (Mueller et al., 1994) and not least also lead to an improved bioavailability (Julianto et
al., 2000; Porter et al.,2008; Trull et al., 1994; Tuleu et al., 2004). SEDDS/SNEDDS
preconcentrates are isotropic mixtures of oils, surfactants, and co-solvents (Mllertz et al.,2010;

310

Pouton and Porter 2008; Small1968). Development of SNEDDS has received most attention in
recent years and will therefore be the focus of this section. The lipid part of the SNEDDS forms

Page 14 of 45

15
the core of the emulsion droplet; hydrophilic surfactants enable the spontaneous emulsifying
process upon dispersing the preconcentrates in aqueous media under mild agitation by forming a
fluidal interfacial film between oil and water phases (Gershanik et al.,2000; Pouton et al.,2008;
Small 1968). Lipophilic surfactants are employed in SNEDDS to ease the emulsification process.

ip
t

315

Addition of co-solvent can lead to a faster dispersion of preconcentrates to nano-emulsions

cr

(Craig et al., 1993; Constantinides 1995; Gershanik et al.,2000; Wakerly et al., 1986). The

preconcentrate and solubilising them into the GI fluids.

an

320

us

excipients may play individual or synergistic roles of dissolving lipophilic compounds in the

Several SNEDDS containing poorly water-soluble drugs have been developed and evaluated for

their in vivo performance (Date and Nagarsenker 2007; Dixit and Nagarsenker 2008b; Dixit and

Nagarsenker 2008a; Wang et al., 2009). Development of SNEDDS is often driven by empiric,

325

te

pseudo-ternary diagrams and solubility of drugs, and is often lacking a systematic approach for
evaluating impact of excipients on the performance of formulations as well as the fate of drugs.

Ac
ce
p

DoE enables evaluating effects of multiple factors, alone or in combination. A number of recent
publications have shown the advantages of DoE in the optimization of SEDDS/SNEDDS
(Basalious et al., 2010; Furlanetto et al., 2011; Holm et al.,2006a; Liu et al., 2010; Taha et al.,
2005). The utilisation of DoE may also improve the understanding on the influence of different
330

excipients and their interactions, as well as predicting optimized formulations with the desired
profile (Holm et al.,2006a). Recently, Ren et al. investigated the influence of different excipients
and their interactions on formulation characteristics of SNEDDS using DoE for a model drug
fenofibrate, and found that the most significant factor in influencing the droplet size of
nanoemulsions upon dispersing SNEDDS preconcentrate was the co-surfactant MaisineTM 35-1,

Page 15 of 45

16
335

which functions as co-surfactant to improve the emulsification of oil, meanwhile it also works as
the oil phase and results in larger droplets (Ren et al.,2012). A significant reduction in droplet

surface activity of fenofibrate and improved self-emulsifying process.

The fate of drugs during the transit in the GI tract may be affected by many factors. As

cr

340

ip
t

size was interestingly observed when fenofibrate was loaded in the vehicles, probably due to the

us

mentioned above, fine emulsions are formed upon dispersing SNEDDS preconcentrates in the GI
fluids, which facilitates lipid hydrolysis by co-lipase dependent pancreatic lipase on the oil/water

an

interface (Chapus et al., 1976; Cordle and Lowe 1998; Smriva and Desnuelle 1979). The
lipolytic products together with bile salts and phospholipids form micelles and other colloidal
structures (Kleberg et al.,2010a; Mllertz et al.,2012). If the solubilisation capacity of micelles is

345

lower than that of the initial fine emulsions, drug compounds may precipitate in the GI tract. The

te

importance of the vesicular species for their drug solubilisation capacity has been investigated by
Porter et al. (2004). They found that dilution with buffer alone resulted in relatively little drug

350

Ac
ce
p

precipitation, whereas dilution with a bile salt solution resulted in extensive precipitation,
presumably resulting from solubilisation of the vesicular lipids by the additional bile salt and a
structural change to a micellar system, which has a much lower capacity for drug solubilization.
For both cinnarizine and danazol a continuous recipitation of drugs was observed during the in
vitro lipolysis process (Sassene et al.,2010; Larsen et al.,2011). However, cinnarizine
precipitated amorphous (Figure 4), which resulted in a fast redissolution (Larsen et al.,2011). For
355

this reason it was speculated that the physical form of drug precipitates during digestion might be
important for the bioavailability of the drug.

Page 16 of 45

17
Recently, Thomas et al. studied supersaturated SNEDDS containing halofantrine (Thomas et al.,
2012a) and simvastatin (Thomas et al.,2012c). When the concentration of halofantrine in the
360

SNEDDS preconcentrate was 150% of the equilibrium solubility, drug precipitation occurred

ip
t

very fast during in vitro lipolysis. However, subsequent dissolution studies of the precipitates
demonstrated enhanced dissolution of halofantrine, which was suggested to be due to an

cr

amorphous form (Thomas et al.,2012a). The enhanced dissolution of the amorphous halofantrine

365

us

was also reflected in vivo where two capsules of conventional SNEDDS were required to achieve
similar AUC and Cmax as obtained after dosing of a single capsule of supersaturated SNEDSS

an

(Thomas et al.,2012a). Therefore drug precipitations are not necessarily a problem during the
dispersion of SNEDDS, but more studies need to be performed in this area in order to get a better

understanding on the phenomena and the mechanism. The stability of supersaturated SNEDDS

and bioavailability.

te

370

should also be considered and addressed for the strategy to be useful in improving absorption

Ac
ce
p

The increased interest on SEDDS/SNEDDS is closely related with the uniformed and
reproducible oral bioavailability of drugs in both the fasted and fed state. DoE can be used in the
optimisation of SEDDS/SNEDDS to simplify the optimisation process and predict the optimised
375

formulations. Supersaturated SNEDDS may be used to improve the bioavailability of poorly


water-soluble drugs, but further studies are required to clarify and confirm the hypothesis of the
physical form of drug precipitates during digestion is important for the bioavailability of the drug.

4.4. Transformation of liquid lipid systems into solid oral dosage forms

Page 17 of 45

18
380

In order to achieve better physicochemical stabilities of LBDDS and ease commercialisation, a


lot of efforts have been devoted to transformation of liquid lipid systems into solid oral dosage
forms. The oldest solidification techniques include spray drying, lyophilisation, and rotary

ip
t

evaporation. Dry emulsions containing poorly water-soluble drugs have been prepared by drying
o/w emulsions containing a solid carrier, which encapsulates the dispersed lipid phase when the
aqueous phase is removed by spray drying (Christensen et al., 2001; Dollo et al., 2003; Takeuchi

cr

385

us

et al., 1991), by lyophilisation (Corveleyn and Remon 1998), or by rotary evaporation (Porter et
al., 1996). O/w emulsions can be reconstituted from dry emulsions upon addition of an aqueous

an

phase, in vivo or in vitro. The ratio of solid carrier to lipid phase influences the reconstitution
properties (Christensen et al.,2001). With the development of lipid-based formulations, spray
drying technique has also been used to solidify other kind of lipid-based formulations, such as

390

SNEDDS. More recent advances in the study of solid SNEDDS with focus on solidification

te

techniques have been summarized by Tang et al. (2008).

395

Ac
ce
p

Yi et al. developed a solid SNEDDS by dispersing SNEDDS preconcentrates containing


nimodipine in a dextran solution followed by spray drying; no difference was observed in the
droplet size of the reconstituted emulsions from liquid and solid SNEDDS (Yi et al., 2008). The
oral bioavailability of nimodipine was investigated in rabbits and similar AUC and Cmax values
were observed from both SNEDDS, but the maximum absorption was retarded by the solid
SNEDDS (Yi et al.,2008). Dixit and Nagarsenker (2008c) studied self-nanoemulsifying granules
400

containing ezetimibe, a cholesterol absorption inhibitor, by mixing SNEDDS preconcentrates


with colloidal silicon dioxide, followed with drying in a desiccators. Dissolution studies revealed
two to three fold increases in the dissolution rate compared to plain drug, in vivo evaluation in

Page 18 of 45

19
rats also showed significant decrease in the total cholesterol levels as compared to positive
control, signifying the potential of such kind of formulation in improving delivery of lipophilic
405

drugs (Dixit et al.,2008c). Recently, Sander and Holm investigated the possibility to solidify

ip
t

SNEDDS using liquid loadable tablets (LLT), which were prepared by direct compression of
porous magnesium aluminometasilicate; SNEDDS containing cyclosporine were loaded by a

cr

simple absorption method (Sander and Holm 2009). The in vivo study in dogs showed similar

410

us

bioavailability of cyclosporine from SNEDDS containing capsules and SNEDDS loaded LLT,
when LLT contained a tablet disintegrant (Sander et al.,2009). Recently, Van Speybroeck et al.

an

(2012) studied the effect of solidification of SEDDS on drug release in vitro and absorption in
vivo when SEDDS were adsorbed onto Neusilin US2 powder. They found that the release of

danazol during in vitro lipolysis was 35% lower for the adsorbed formulation, and the

formulations due to incomplete desorption of SEDDS. Therefore selection of lipid excipients,

te

415

bioavailability of danazol from solidified formulation in rats was 50% lower than the liquid

Ac
ce
p

solid carriers as well as solidification process should be carefully considered.

Effects of silica encapsulation has been investigated by Takeuchi et al. (1991), who compared
the oral bioavailability of vitamin E acetate from a silica encapsulated drug emulsion, a
420

conventional oil, and a liquid emulsion. No significant differences were observed in the
pharmacokinetic parameters (AUC, Cmax, or Tmax). More recently silica-lipid hybrid (SLH)
microcapsule systems have been used to improve the bioavailability of poorly water-soluble
drugs (Simovic et al., 2010; Tan et al., 2009; Tan et al., 2011). SLH microcapsules composed of
MCT, lecithin and silica nanoparticles resulted in improved AUC (250-270 min g/ml) of

425

celecoxib compared to a lipid solution (170 min g/ml) and an o/w emulsion (197 min g/ml),

Page 19 of 45

20
that was suggested to be a reflection of dissolution enhancement (Tan et al.,2009). Quantification
of the lipase-mediated digestion kinetics in vitro showed the lag phase duration of 5 s, 7-10 s,
and 18-23 s for lipid emulsion, SLH, and lipid solution, respectively; the initial digestion rate

430

ip
t

constant was highest for lipid emulsions (1.9 min-1), followed with SLH (1.2-1.5 min-1), and
lowest for lipid solution (0.5-0.8 min-1) (Tan et al., 2010). This confirms the importance of

cr

emulsification on lipid digestion. Details about mesoporous silicon and assembling nanoparticle

us

coatings can be found in the recent reviews (Prestidge et al., 2007; Simovic et al., 2012). Salonen
and Lehto (2008) also described the fabrication and chemical modifications of porous silicon for

435

an

biomedical applications. Tanaka et al. (2010) studied the safety of nanoporous silicon carriers in
vivo followed single and multiple dose intravenous administrations in mice and claimed that they

provided evidence of safe intravenous administration.

te

Solidification of liquid lipid systems may improve the physiochemical stabilities of LBDDS. The
selection of the solidification techniques and solid carriers is important to ensure sufficient
desorption of drugs as well as lipid excipients from solid carriers. More safety studies on

Ac
ce
p

440

solidifying carriers should also be performed.

5. In vitro and in vivo effects: comparison of dosage forms and lipid excipients
Both selection of lipid excipients and dosage forms can be important for drug solubilisation and
445

absorption. However, only a limited number of studies have compared the performance of
different LBDDS in vivo and/or in vitro. Han et al. (2009) compared the performance of lipid
solution and sub-microemulsion of a lipophilic drug anethol trithione both in vivo and in vitro,
and the formulations were prepared using SCT, MCT, and LCT. Sub-microemulsion

Page 20 of 45

21
formulations were digested more rapidly in vitro than oil solutions, and the solubilisation
450

capacity during lipolysis was in the order of SCT<MCT<LCT. All LBDDS enhanced drug
absorption compared to an aqueous suspension, but the drug absorption was better from

cr

ip
t

emulsions than from lipid solutions when LCT was used (Han et al.,2009).

455

us

Nielsen et al. (2008) studied the bioavailability of probucol from lipid solutions and
SEDDS/SNEDDS in fasted and fed minipigs and found higher bioavailability of probucol from

an

SEDDS/SNEDDS than from oil solutions, but the difference was not significant. In vitro studies
on the same formulations showed that the release rate and extent of release of probucol from the

oil solution were significantly lower than that from SEDDS/SNEDDS, and the rank order of
probucol released (SEDDS/SNEDDS> oil solution) was similar to the rank order of
bioavailability from the in vivo study (Fatouros et al., 2008). Grove et al. (2007) also studied the

460

te

bioavailability of seocalcitol in minipigs; two lipid-based formulations were investigated, one

Ac
ce
p

was lipid solution and the other was SNEDDS. The study was carried out both in fasted and fed
state, similar seocalcitol bioavailability was observed from the two different formulations in both
cases (Grove et al.,2007).
465

Kuentz et al (2007) investigated the role of lipid formulation parameters on the oral
bioavailability of the compound CDA (log P=8.9) in rats using a multivariate statistical method.
They used different lipid excipients, including MCT, LCT, mixture of DG and MG, for diverse
formulations including SEDD, SNEDDS and lipid solutions. They found that SEDDS, especially
470

SNEDDS, were most effective in pushing the exposure of the compound CDA, there was a
tendency (not significant) toward lower exposure when LCT was used in the formulation

Page 21 of 45

22
(Kuentz et al.,2007). In a recent in vitro lipolysis study in biorelevant media, different LFCS type
formulations were used to investigate the influence of lipid digestion on the distribution of
fenofibrate between the aqueous phase and the pellet phase (Mohsin2012). As expected type II
and type IIIA formulations were rapidly digested in the initial stages because of the larger

ip
t

475

surface area of emulsions. However, a high concentration of surfactant used in type IIIB and IV

cr

systems lowered the rate of digestion. Type I formulations presented comparably higher amount

us

of fenofibrate in aqueous phase after 30 min lipolysis, even though it had lower initial digestion
rate. It was concluded that maintaining drug in aqueous phase depended on the solubilisation of
drug by mixed bile salt micelles and lipolytic products (Mohsin2012). It should be noted,

an

480

however, that no comparative in vivo studies were carried out.

In vitro lipolysis model has been increasingly used in mimicking the enzymatic degradation and

of the in vitro model in understanding of how LBDDS promote drug delivery, it requires more

Ac
ce
p

485

te

monitoring drug solubility during the lipid digestion process. In order to broaden the utilisation

studies on better characterizations of lipid degradation products in vitro and correlation studies of
in vitro and in vivo.

6. Summary
490

LBDDS have great potentials for improving oral bioavailability of poorly water-soluble drugs,
especially lipophilic drugs. Rational design of lipid-based formulations can be achieved by
utilisation of DoE, which can also be used in prediction of optimised formulations. It is possible
to target lymphatic transport of drugs by selection of lipid excipients containing long-chain fatty
acids for lipophilic drugs with good solubility in lipids. Drug precipitation during dispersing of

Page 22 of 45

23
495

lipid-based formulations in the GI fluids may affect bioavailability, possibly depending on the
polymorphic form of precipitated drugs. Supersaturated SNEDDS may be utilised improving
bioavailabilities, but their stability should be carefully considered. The in vitro lipolysis model is

ip
t

a useful tool in characterisation of LBDDS and may be used for prediction of drug solubility in
the GI fluids during dispersing and lipid digestion processes. However, a better understanding of
the mechanisms behind the absorption of drugs from LBDDS, including the important

cr

500

us

parameters in the in vitro lipolysis, is needed in order to enable a rational optimisation of

an

LBDDS with high and reproducible performance.

References
505

Abrams, L.S., Weintraub, H.S., Patrick, J.E., Mcguire, J.L., 1978. Comparative bioavailability of

a lipophilic steroid. J. Pharm. Sci. 67, 1287-1290.

te

Amidon, G.L., Lennernas, H., Shah, V.P., Crison, J.R., 1995. A theoretical basis for a
biopharmaceutic drug classification - the correlation of in-vitro drug product dissolution
and in-vivo bioavailability. Pharm. Res. 12, 413-420.

Ac
ce
p

510

Armand, M., 2007. Lipases and lipolysis in the human digestive tract: where do we stand? Curr.
Opin. Clin. Nutr. & Metab. Care 10, 156-164.
Basalious, E.B., Shawky, N., Badr-Eldin, S.M., 2010. SNEDDS containing bioenhancers for
improvement of dissolution and oral absorption of lacidipine. I: Development and
515

optimization. Int. J. Pharm. 391, 203-211.


Caliph, S.M., Charman, W.N., Porter, C.J., 2000. Effect of short-, medium-, and long-chain fatty
acid-based vehicles on the absolute oral bioavailability and intestinal lymphatic transport

Page 23 of 45

24
of halofantrine and assessment of mass balance in lymph-cannulated and non-cannulated
rats. J. Pharm. Sci. 89, 1073-1084.
520

Carrigan, P.J., Bates, T.R., 1973. Biopharmaceutics of drugs administered in lipid-containing

ip
t

dosage forms .1. Gi-absorption of griseofulvin from an oil-in-water emulsion in rat. J.


Pharm. Sci. 62, 1476-1479.

cr

Cast, J., Hamilton, R.J., 1999. in: Hamilton R.J. and Cast J. (Eds) , Spectral Peroperties of

525

us

Lipids. CRC Press LLC, Boca Raton, pp. 1-19.

Castera, A., 1995. in: Sebedio J.-L. and Perkins E.G. (Eds) , New Trends in Lipid and

an

Lipoprotein Analyses. AOCS Press, Champaign, pp. 10-16.

Chakrabarti, S., Belpaire, F.M., 1978. Bioavailability of phenytoin in lipid containing dosage

forms in rats. J. Pharm. Pharmacol. 30, 330-331.

delivery of drugs with poor bioavailability. Eur. J. Pharm. Biopharm. 73, 1-15.

te

530

Chakraborty, S., Shukla, D., Mishra, B., Singh, S., 2009. Lipid - An emerging platform for oral

Chapus, C., Smriva, M., Bovier-Lapierre, C., Desnuelle, P., 1976. Mechanism of pancreatic

Ac
ce
p

lipase action. 1. Interfacial activation of pancreatic lipase. Biochem. 15, 4980-4987.


Charman, W.N.A., Stella, V.J., 1986a. Effects of lipid class and lipid vehicle volume on the
intestinal lymphatic transport of DDT. Int. J. Pharm. 33, 165-172.
535

Charman, W.N.A., Stella, V.J., 1986b. Estimating the maximal potential for intestinal lymphatic
transport of lipophilic drug molecules. Int. J. Pharm. 34, 175-178.
Christensen, J.O., Schultz, K., Mollgaard, B., Kristensen, H.G., Mullertz, A., 2004.
Solubilisation of poorly water-soluble drugs during in vitro lipolysis of medium- and
long-chain triacylglycerols. Eur. J. Pharm. Sci. 23, 287-296.

Page 24 of 45

25
540

Christensen, K.L., Pedersen, G.P., Kristensen, H.G., 2001. Preparation of redispersible dry
emulsions by spray drying. Int. J. Pharm. 212, 187-194.
Christie, W.W., 1985. Rapid separation and quantification of lipid classes by high performance

ip
t

liquid chromatography and mass (light-scattering) detection. J. Lipid Res. 26, 507-512.
Christophe, A.B., 2004. Digestion and absorption of glycerides, in: Katsuragi Y., Yasukawa T.,
Matsuo N., Flickinger B.D., Tokimitsu I., and Matlock M.G. (Eds) , Diacylglycerol Oil.

cr

545

us

AOCS Press, Champaign, pp. 16-29.

Constantinides, P.P., 1995. Lipid microemulsions for improving drug dissolution and oral

an

absorption: physical and biopharmaceutical aspects. Pharm. Res. 12, 1561-1572.


Cordle, R.A., Lowe, M.E., 1998. The hydrophobic surface of colipase influences lipase activity
at an oil-water interface. J. Lipid Res. 39, 1759-1767.

550

Corveleyn, S., Remon, J.P., 1998. Formulation of a lyophilized dry emulsion tablet for the

te

delivery of poorly soluble drugs. Int. J. Pharm. 166, 65-74.


Craig, D.Q.M., Lievens, H.S.R., Pitt, K.G., Storey, D.E., 1993. An investigation into the

555

Ac
ce
p

physico-chemical properties of self-emulsifying systems using low frequency dielectric


spectroscopy, surface tension measurements and particle size analysis. Int. J. Pharm. 96,
147-155.

Dahan, A., Hoffman, A., 2007. The effect of different lipid based formulations on the oral
absorption of lipophilic drugs: The ability of in vitro lipolysis and consecutive ex vivo
intestinal permeability data to predict in vivo bioavailability in rats. Eur. J. Pharm.
560

Biopharm. 67, 96-105.


Date, A.A., Nagarsenker, M.S., 2007. Design and evaluation of self-nanoemulsifying drug
delivery systems (SNEDDS) for cefpodoxime proxetil. Int. J. Pharm. 329, 166-172.

Page 25 of 45

26
Dixit, R.P., Nagarsenker, M.S., 2008a. Formulation and in vivo evaluation of selfnanoemulsifying granules for oral delivery of a combination of ezetimibe and simvastatin.
565

Drug Develop. Ind. Pharm. 34, 1285-1296.

optimization and evaluation. Eur. J. Pharm. Sci. 35, 183-192.

ip
t

Dixit, R.P., Nagarsenker, M.S., 2008b. Self-nanoemulsifying granules of ezetimibe: Design,

cr

Dixit, R.P., Nagarsenker, M.S., 2008c. Self-nanoemulsifying granules of ezetimibe: Design,

570

us

optimization and evaluation. Eur. J. Pharm. Sci. 35, 183-192.

Dollo, G., Le Corre, P., Guerin, A., Chevanne, F., Burgot, J.L., Leverge, R., 2003. Spray-dried

an

redispersible oil-in-water emulsion to improve oral bioavailability of poorly soluble drugs.


Eur. J. Pharm. Sci. 19, 273-280.

Drewe, J., Meier, R., Vonderscher, J., Kiss, D., Posanski, U., Kissel, T., Gyr, K., 1992.

60-64.

te

575

Enhancement of the oral absorption of cyclosporine in man. Br. J. Clin. Pharmacol. 34,

Fatouros, D.G., Deen, G.R., Arleth, L., Bergenstahl, B., Nielsen, F.S., Pedersen, J.S., Mullertz,

Ac
ce
p

A., 2007. Structural development of self nano emulsifying drug delivery systems
(SNEDDS) during In vitro lipid digestion monitored by small-angle x-ray scattering.
Pharm. Res. 24, 1844-1853.
580

Fatouros, D., Nielsen, F., Douroumis, D., Hadjileontiadis, L., Mllertz, A., 2008. In vitro-in vivo
correlations of self-emulsifying drug delivery systems combining the dynamic lipolysis
model and neuro-fuzzy networks. Eur. J. Pharm. Biopharm. 69, 887-898.
Fricker, G. et al., 2010. Phospholipids and lipid-based formulations in oral drug delivery. Pharm.
Res. 27, 1469-1486.

Page 26 of 45

27
585

Furlanetto, S., Cirri, M., Piepel, G., Mennini, N., Mura, P., 2011. Mixture experiment methods in
the development and optimization of microemulsion formulations. J. Pharm. Biomed.
Anal. 55, 610-617.

of lipophilic drugs. Eur. J. Pharm. Biopharm. 50, 179-188.

Gershkovich, P., Qadri, B., Yacovan, A., Amselem, S., Hoffman, A., 2007. Different impacts of

cr

590

ip
t

Gershanik, T., Benita, S., 2000. Self-dispersing lipid formulations for improving oral absorption

us

intestinal lymphatic transport on the oral bioavailability of structurally similar synthetic


lipophilic cannabinoids: dexanabinol and PRS-211,220. Eur. J. Pharm. Sci. 31, 298-305.

an

Grove, M., Mullertz, A., Pedersen, G.P., Nielsen, J.L., 2007. Bioavailability of seocalcitol III.
Administration of lipid-based formulations to minipigs in the fasted and fed state. Eur. J.
Pharm. Sci. 31, 8-15.

595

Grove, M., Pedersen, G.P., Nielsen, J.L., Mullertz, A., 2005. Bioavailability of seocalcitol I:

te

Relating solubility in biorelevant media with oral bioavailability in rats - Effect of


medium and long chain triglycerides. J. Pharm. Sci. 94, 1830-1838.

600

Ac
ce
p

Gursoy, R.N., Benita, S., 2004. Self-emulsifying drug delivery systems (SEDDS) for improved
oral delivery of lipophilic drugs. Biomed. Pharmacother. 58, 173-182.
Hamosh, M., 1979. Review - Fat digestion in the newborn - role of lingual lipase and preduodenal digestion. Pediatr. Res. 13, 615-622.
Hamosh, M., Scow, R.O., 1973. Lingual lipase and its role in digestion of dietary lipid. J. Clin.
Invest. 52, 88-95.
605

Han, S.F., Yao, T.T., Zhang, X.X., Gan, L., Zhu, C.L., Yua, H.Z., Gan, Y., 2009. Lipid-based
formulations to enhance oral bioavailability of the poorly water-soluble drug anethol
trithione: Effects of lipid composition and formulation. Int. J. Pharm. 379, 18-24.

Page 27 of 45

28
Hargrove, J.T., Maxson, W.S., Wentz, A.C., 1989. Absorption of oral progesterone is influenced
by vehicle and particle-size. Am. J. Obstet. Gynecol. 161, 948-951.
610

Hauss D.J., 2007. Oral lipid-based formulations. Adv. Drug Delivery Rev. 59, 667-676.

ip
t

Heinz, E., 2012. in: Christie W.W. (Ed.) , Advances In Lipid Methodology - Three. The Oily
Press, Dundee, pp. 211-332.

cr

Holm, R., Jensen, I.H.M., Sonnergaard, J., 2006a. Optimization of self-microemulsifying drug

615

Drug Develop. Ind. Pharm. 32, 1025-1032.

us

delivery systems (SMEDDS) using a D-optimal design and the desirability function.

an

Holm, R., Porsgaard, T., Porter, C.J.H., Hoy, C.E., Edwards, G.A., Mullertz, A., Kristensen,
H.G., Charman, W.N., 2006b. Lymphatic fatty acids in canine with pharmaceutical

formulations containing structured triacylglycerols. Eur. J. Lipid Sci. Technol. 108, 714-

Holm, R., Porter, C.J.H., Edwards, G.A., Mullertz, A., Kristensen, H.G., Charman, W.N., 2003.

te

620

722.

Examination of oral absorption and lymphatic transport of halofantrine in a triple-

Ac
ce
p

cannulated canine model after administration in self-microemulsifying drug delivery


systems (SMEDDS) containing structured triglycerides. Eur. J. Pharm. Sci. 120, 91-97.
Julianto, T., Yuen, K.H., Noor, A.M., 2000. Improved bioavailability of vitamin E with a self
625

emulsifying formulation. Int. J. Pharm. 200, 53-57.


Kalogeris, T.J., Monroe, F., DeMichele, S.J., Tso, P., 1996. Intestinal synthesis and lymphatic
secretion of apolipoprotein A-IV vary with chain length of intestinally infused fatty acids
in rats. J. Nutr. 126, 2720-2729.

Page 28 of 45

29
Kaukonen, A.M., Boyd, B.J., Charman, W.N., Porter, C.J.H., 2004a. Drug solubilization
630

behavior during in vitro digestion of suspension formulations of poorly water-soluble


drugs in triglyceride lipids. Pharm. Res. 21, 254-260.

ip
t

Kaukonen, A.M., Boyd, B.J., Porter, C.J.H., Charman, W.N., 2004b. Drug solubilization

behavior during in vitro digestion of simple triglyceride lipid solution formulations.

Kleberg, K., Jacobsen, F., Fatouros, D.G., Mullertz, A., 2010a. Biorelevant media simulating fed

us

635

cr

Pharm. Res. 21, 245-253.

state intestinal fluids: colloid phase characterization and impact on solubilization capacity.

an

J. Pharm. Sci. 99, 3522-3532.

Kleberg, K., Jacobsen, J., Mullertz, A., 2010b. Characterising the behaviour of poorly water

and transport studies. J. Pharm. Pharmacol. 62, 1656-1668.

640

soluble drugs in the intestine: application of biorelevant media for solubility, dissolution

te

Kohli, K., Chopra, S., Dhar, D., Arora, S., Khar, R.K., 2010. Self-emulsifying drug delivery
systems: an approach to enhance oral bioavailability. Drug Discov. Today 15, 958-965.

Ac
ce
p

Kristensen, J., Jorgensen, H., Mu, H., 2006. Diacylglycerol oil does not affect portal vein
transport of nonesterified fatty acids but decreases the postprandial plasma lipid response
645

in catheterized pigs. J Nutr. 136, 1800-1805.


Kuentz, M., 2011. Oral self-emulsifying drug delivery systems, from biopharmaceutical to
technical formulation aspects. J. Drug Deliv. Sci. Technol. 21, 17-26.
Kuentz, M., Wyttenbach, N., Kuhlmann, O., 2007. Application of a statistical method to the
absorption of a new model drug from micellar and lipid formulations - Evaluation of

650

qualitative excipient effects. Pharm. Develop. Tech. 12, 275-283.

Page 29 of 45

30
Larsen, A., Holm, R., Pedersen, M.L., Mullertz, A., 2008. Lipid-based formulations for danazol
containing a digestible surfactant, labrafil m2125cs: in vivo bioavailability and dynamic
in vitro lipolysis. Pharm. Res. 25, 2769-2777.

655

ip
t

Larsen, A.T., Sassene, P., Mullertz, A., 2011. In vitro lipolysis models as a tool for the

characterization of oral lipid and surfactant based drug delivery systems. Int. J. Pharm.

cr

417, 245-255.

us

Lipinski, C.A., 2000. Drug-like properties and the causes of poor solubility and poor
permeability. J. Pharmacol. Toxicol. Methods 44, 235-249.

660

an

Lipinski, C.A., Lombardo, F., Dominy, B.W., Feeney, P.J., 1997. Experimental and
computational approaches to estimate solubility and permeability in drug discovery and

development settings. Adv. Drug Delivery Rev. 23, 3-25.

Liu, Y., Chen, Z.Q., Zhang, X., Feng, N.P., Zhao, J.H., Wu, S., Tan, R., 2010. An improved

te

formulation screening and optimization method applied to the development of a selfmicroemulsifying drug delivery system. Chem. Pharm. Bull. 58, 16-22.
Moffett D.F., Moffett S.B., Schauf C.L., 1993. Human physiology, foundations & frontiers,

Ac
ce
p

665

London.

Mohsin, K., 2012. Design of lipid-based formulations for oral administration of poorly watersoluble drug fenofibrate: effects of digestion. AAPS Pharm. Sci. Tech. 13, 637-646.
Mu, H., 2005. Analysis of lipids by new hyphenated techniques, in: Akoh C.C. and Lai O.M.
670

(Eds) , Healthful Lipids. AOCS Press, Champaign, pp. 78-98.


Mu, H., Hy, C.-E., 2004. The digestion of dietary triacylglycerols. Prog. Lipid Res. 43, 105-133.
Mu, H., Kalo, P., Xu, X., Hy, C.-E., 2000a. Chromatographic methods in the monitoring of
lipase-catalyzed interesterification. Eur. J. Lipid Sci. Technol. 102, 202-211.

Page 30 of 45

31
Mu, H., Hy, C.-E., 2000b. Effect of medium-chain fatty acids on lymphatic absorption of
675

essential fatty acids in specific structured lipids. Lipids 35, 83-89.


Mu, H., Porsgaard, T., 2005. The metabolism of structured triacylglycerols. Prog. Lipid Res. 44,

ip
t

430-448.

Mu, H., Hoy, C.E., 2001. Intestinal absorption of specific structured triacylglycerols. J. Lipid

Mueller, E.A., Kovarik, J.M., Vanbree, J.B., Grevel, J., Lucker, P.W., Kutz, K., 1994. Influence

us

680

cr

Res. 42, 792-798.

of a fat-rich meal on the pharmacokinetics of a new oral formulation of cyclosporine in a

an

crossover comparison with the market formulations. Pharm. Res. 11, 151-155.
Mllertz, A., Fatouros, D.G., Smith, J.R., Vertzoni, M., Reppas, C., 2012. Insights into

cryo-transmission electron microscopy ex vivo. Mol. Pharm. 9, 237-247.

685

intermediate phases of human intestinal fluids visualized by atomic force microscopy and

te

Mllertz, A., Ogbonna, A., Ren, S., Rades, T., 2010. New perspectives on lipid and surfactant
based drug delivery systems for oral delivery of poorly soluble drugs. J. Pharm.

Ac
ce
p

Pharmacol. 62, 1622-1636.

Nielsen, F.S., Petersen, K.B., Mullertz, A., 2008. Bioavailability of probucol from lipid and
690

surfactant based formulations in minipigs: Influence of droplet size and dietary state. Eur.
J. Pharm. Biopharm. 69, 553-562.
Pitzalis, P., Monduzzi, M., Krog, N., Larsson, H., Ljusberg-Wahren, H., Nylander, T., 2000.
Characterization of the liquid-crystalline phases in the glycerol monooleate/diglycerol
monooleate/water system. Langmuir 16, 6358-6365.

695

Porter, C.J., Trevaskis, N.L., Charman, W.N., 2007. Lipids and lipid-based formulations:
optimizing the oral delivery of lipophilic drugs. Nat. Rev Drug Discov. 6, 231-248.

Page 31 of 45

32
Porter, C.J.H., Charman, S.A., Williams, R.D., Bakalova, M.V., Charman, W.N., 1996.
Evaluation of emulsifiable glasses for the oral administration of cyclosporin in beagle
dogs. Int. J. Pharm. 141, 227-237.
Porter, C.J.H., Kaukonen, A.M., Taillardat-Bertschinger, A., Boyd, B.J., O'Connor, J.M.,

ip
t

700

Edwards, G.A., Charman, W.N., 2004. Use of in vitro lipid digestion data to explain the

cr

in vivo performance of triglyceride-based oral lipid formulations of poorly water-soluble

us

drugs: Studies with halofantrine. J. Pharm. Sci. 93, 1110-1121.

Porter, C., Pouton, C.W., Cuine, J.F., Charman, W.N., 2008. Enhancing intestinal drug
solubilisation using lipid-based delivery systems. Adv. Drug Delivery Rev. 60, 673-691.

an

705

Pouton, C.W., 2000. Lipid formulations for oral administration of drugs: non-emulsifying, self-

emulsifying and 'self-microemulsifying' drug delivery systems. Eur. J. Pharm. Sci. 11,

S93-S98.

710

te

Pouton, C.W., 2006. Formulation of poorly water-soluble drugs for oral administration:
physicochemical and physiological issues and the lipid formulation classification system.

Ac
ce
p

Eur. J. Pharm. Sci. 29, 278-287.

Pouton, C., Porter, CJ., 2008. Formulation of lipid-based delivery systems for oral administration:
materials, methods and strategies. Adv. Drug Delivery Rev. 60, 625-637.
Prestidge, C.A., Barnes, T.J., Lau, C.H., Barnett, C., Loni, A., Canham, L., 2007. Mesoporous
715

silicon: a platform for the delivery of therapeutics. Expert Opin. Drug Deliv. 4, 101-110.
Rahman, M.A., Harwansh, R., Mirza, M.A., Hussain, S., Hussain, A., 2011. Oral lipid based
drug delivery system (LBDDS): formulation, characterization and application: a review.
Curr. Drug Deliv. 8, 330-345.

Page 32 of 45

33
Rane, S.S., Anderson, B.D., 2008. What determines drug solubility in lipid vehicles: Is it
720

predictable? Adv. Drug Delivery Rev. 60, 638-656.


Ren, S., Mu, H.L., Alchaer, F., Chtatou, A., Mllertz, A., 2012. Optimization of self-nano-

ip
t

emulsifying drug delivery system for poorly water soluble drug using response surface
methodology. Drug Develop. Ind. Pharm., e-adead published Aug. 2012.

ingredients. AAPS Pharm. Sci. Tech. 13, 1103-1109.

us

725

cr

Sacchetti, M., Nejati, E., 2012. Prediction of drug solubility in lipid mixtures from the individual

Salonen, J., Lehto, V.P., 2008. Fabrication and chemical surface modification of mesoporous

an

silicon for biomedical applications. Chem. Eng. J. 137, 162-172.

Sander, C., Holm, P., 2009. Porous magnesium aluminometasilicate tablets as carrier of a

Sassene, P.J., Knopp, M.M., Hesselkilde, J.Z., Koradia, V., Larsen, A., Rades, T., Mullertz, A.,

730

cyclosporine self-emulsifying formulation. AAPS Pharm. Sci. Tech. 10, 1388-1395.

te

2010. Precipitation of a poorly soluble model drug during in vitro lipolysis:


characterization and dissolution of the precipitate. J. Pharm. Sci. 99, 4982-4991.

Ac
ce
p

Smriva, M., Desnuelle, P., 1979. Pancreatic lipase and colipase. an example of heterogeneous
biocatalysis. Adv. Enzymol. 48, 319-370.
735

Shimada, A., Ohashi, K., 2003. Interfacial and emulsifying properties of diacylglycerol. Food Sci.
Tech. Res. 9, 142-147.

Shukla, D., Chakraborty, S., Singh, S., Mishra, B., 2011. Lipid-based oral multiparticulate
formulations - advantages, technological advances and industrial applications. Expert
Opin. Drug Deliv. 8, 207-224.
740

Simovic, S., Barnes, T.J., Tan, A., Prestidge, C.A., 2012. Assembling nanoparticle coatings to
improve the drug delivery performance of lipid based colloids. Nanoscale 4, 1220-1230.

Page 33 of 45

34
Simovic, S., Hui, H., Song, Y.M., Davey, A.K., Rades, T., Prestidge, C.A., 2010. An oral
delivery system for indomethicin engineered from cationic lipid emulsions and silica
nanoparticles. J. Control. Release 143, 367-373.
Singh, A., Worku, Z.A., Van den Mooter, G., 2011. Oral formulation strategies to improve

ip
t

745

solubility of poorly water-soluble drugs. Expert Opin. Drug Deliv. 8, 1361-1378.

cr

Singh, B., Bandopadhyay, S., Kapil, R., Singh, R., Katare, O.P., 2009a. Self-emulsifying drug

Crit. Rev. Ther. Drug Carrier Syst. 26, 427-521.

Singh, H., Ye, A.Q., Horne, D., 2009b. Structuring food emulsions in the gastrointestinal tract to

an

750

us

delivery systems (SEDDS): formulation development, characterization, and applications.

modify lipid digestion. Prog. Lipid Res. 48, 92-100.

Small, D.M., 1968. A classification of biologic lipids based upon their interaction in aqueous

systems. J. Am. Oil Chem. Soc. 45, 108-119.

755

te

Smith, L.J., Kaminsky, S., Dsouza, S.W., 1986. Neonatal fat digestion and lingual lipase. Acta
Paediatr. Scand. 75, 913-918.

Ac
ce
p

Strickley, R.G., 2004. Solubilizing excipients in oral and injectable formulations. Pharm. Res. 21,
201-230.

Taha, E.I., Samy, A.M., Kassem, A.A., Khan, M.A., 2005. Response surface methodology for
the development of self-nanoemulsified drug delivery system (SNEDDS) of all-trans760

retinol acetate. Pharm. Dev. Tech. 10, 363-370.


Takeuchi, H., Sasaki, H., Niwa, T., Hino, T., Kawashima, Y., Uesugi, K., Ozawa, H., 1991.
Redispersible dry emulsion system as novel oral dosage form of oily drugs - in vivo
studies in beagle dogs. Chem. Pharm. Bull. 39, 3362-3364.

Page 34 of 45

35
Tan, A., Davey, A.K., Prestidge, C.A., 2011. Silica-lipid hybrid (slh) versus non-lipid
765

formulations for optimising the dose-dependent oral absorption of celecoxib. Pharm. Res.
28, 2273-2287.

ip
t

Tan, A., Simovic, S., Davey, A.K., Rades, T., Boyd, B.J., Prestidge, C.A., 2010. Silica

nanoparticles to control the lipase-mediated digestion of lipid-based oral delivery systems.

Tan, A., Simovic, S., Davey, A.K., Rades, T., Prestidge, C.A., 2009. Silica-lipid hybrid (SLH)

us

770

cr

Mol. Pharm. 7, 522-532.

microcapsules: A novel oral delivery system for poorly soluble drugs. J. Control. Release

an

134, 62-70.

Tanaka, T. et al., 2010. In vivo evaluation of safety of nanoporous silicon carriers following

Tang, B., Cheng, G., Gu, J.C., Xu, C.H., 2008. Development of solid self-emulsifying drug

775

single and multiple dose intravenous administrations in mice. Int. J. Pharm. 402, 190-197.

612.

te

delivery systems: preparation techniques and dosage forms. Drug Discov. Today 13, 606-

Ac
ce
p

Thomas, N., Holm, R., Mllertz, A., Rades, T., 2012a. In vitro and in vivo performance of novel
supersaturated self-nanoemulsifying drug delivery systems (super-SNEDDS). J. Control.
780

Release 160, 25-32.

Thomas, N., Holm, R., Rades, T., Mullertz, A., 2012b. Characterising lipid lipolysis and its
implication in lipid-based formulation development. AAPS J. 14, 860-871.
Thomas, N., Mullertz, A., Graf, A., Rades, T., 2012c. Influence of lipid composition and drug
load on the in vitro performance of self-nanoemulsifying drug delivery systems. J. Pharm.
785

Sci. 101, 1721-1731.

Page 35 of 45

36
Trull, A.K., Tan, K.K.C., Tan, L., Alexander, G.J.M., Jamieson, N.V., 1994. Enhanced
absorption of new oral cyclosporine microemulsion formulation, neoral, in livertransplant recipients with external biliary diversion. Transplant. Proc. 26, 2977-2978.

790

ip
t

Tso, P., 1985. Gastrointestinal digestion and absorption of lipid. Adv. Lipid Res. 21, 143-186.
Tso, P., 1994. Intestinal lipid absorption, in: Johnson L.R. (Ed.) , Physiology of the

cr

Gastrointestinal Tract. Raven Press, New York, pp. 1867-1907.

us

Tuleu, C., Newton, M., Rose, J., Euler, D., Saklatvala, R., Clarke, A., Booth, S., 2004.
Comparative bioavailability study in dogs of a self-emulsifying formulation of

795

an

progesterone presented in a pellet and liquid form compared with an aqueous suspension
of progesterone. J. Pharm. Sci. 93, 1495-1502.

Van Speybroeck, M., Williams, H.D., Nguyen, T.H., Anby, M.U., Porter, C.J.H., Augustijns, P.,

2012. Incomplete desorption of liquid excipients reduces the in vitro and in vivo

te

performance of self-emulsifying drug delivery systems solidified by adsorption onto an


inorganic mesoporous carrier. Mol. Pharm. 9, 2750-2760.
Wakerly, M.G., Pouton, C.W., Meakin, B.J., Morton, F.S., 1986. Self-emulsification of

Ac
ce
p

800

vegetable oil-nonionic surfactant mixtures, in: phenomena in mixed surfactant systems.


American Chemical Society, pp. 242-255.
Wang, L.J., Dong, J.F., Chen, J., Eastoe, J.L., Li, X.F., 2009. Design and optimization of a new
self-nanoemulsifying drug delivery system. J. Colloid Interface Sci. 330, 443-448.
805

Watanabe, H., Tokimitsu, I., 2004. Digestion and absorption of diacylglycerol, in: Katsuragi Y.,
Yasukawa T., Matsuo N., Flickinger B.D., Tokimitsu I., and Matlock M.G. (Eds) ,
Diacylglycerol Oil. AOCS Press, Champaign, pp. 30-45.

Page 36 of 45

37
Yanez, J.A., Wang, S.W.J., Knemeyer, I.W., Wirth, M.A., Alton, K.B., 2011. Intestinal
lymphatic transport for drug delivery. Adv. Drug Delivery Rev. 63, 923-942.
810

Yi, T., Wan, J.L., Xu, H.B., Yang, X.L., 2008. A new solid self-microemulsifying formulation

ip
t

prepared by spray-drying to improve the oral bioavailability of poorly water soluble


drugs. Eur. J. Pharm. Biopharm. 70, 439-444.

cr

Yu, L.X. et al., 2002. Biopharmaceutics classification system: the scientific basis for biowaiver

815

us

extensions. Pharm. Res. 19, 921-925.

Zangenberg, N.H., Mullertz, A., Kristensen, H.G., Hovgaard, L., 2001a. A dynamic in vitro

an

lipolysis model. I. Controlling the rate of lipolysis by continuous addition of calcium. Eur.
J. Pharm. Sci. 14, 115-122.

Zangenberg, N.H., Mullertz, A., Kristensen, H.G., Hovgaard, L., 2001b. A dynamic in vitro

te
Ac
ce
p

820

lipolysis model. II: Evaluation of the model. Eur. J. Pharm. Sci. 14, 237-244.

Page 37 of 45

38
Figure captions:
Figure 1. Simplified digestion process of lipid-based formulations. Redrawn and adapted from
Moffett et al. 1993(Moffett et al., 1993).
Figure 2. In vitro lipolysis and drug precipitation, A) the typical set up of a lipolysis model, B)

ip
t

825

phase separation of lipolysis sample after ultracentrifugation, C) distribution (%) of

cr

fenofibrate in the aqueous phase ( ) and precipitates ( ) upon 3 min of dispersion and

us

30 min of digestion of the SNEDDS preconcentrates in a lipolysis medium. Scattered


plots ( ) indicate the dissolved mass of fenofibrate (mg) in the aqueous phase. The
concentration of cosolvent ethanol in the formulation I, II, and III is 0, 5, and 10%

an

830

respectively. Redrawn and adapted from Larsen et al. 2011 and Ren et al. 2012.

Figure 3. Lymphatic transport of lipids and halofantrine solubilised in lipids. A) Cumulative

lymphatic transport of fatty acids after intragastric administration of structured TG

835

te

8:0/18:2/8:0, 10:0/18:2/10:0, 12:0/18:2/12:0; B) cumulative lymphatic transport of


halofantrine after oral administration in different TG solutions. Redrawn and adapted

Ac
ce
p

from Mu & Hy 2000b and Caliph et al. 2000.

Figure 4. (A) Partition of cinnarizine during in vitro lipolysis in the duodenum step. The black
bars are the percentage of cinnarizine in the aqueous phase and the grey bars show the
percentage of cinnarizine precipitated in the pellet. The curve shows the addition of

840

1.0 M NaOH throughout the lipolysis. (B) Diffractogram of cinnarizine obtained using
XRPD. (a) Crystalline cinnarizine, (b) Pellet from in vitro lipolysis containing
precipitated cinnarizine, (c) blank pellet, (d) blank pellet spiked with crystalline
cinnarizine (Adapted from Larsen et al. 2011 and Sassene et al., 2010).

Page 38 of 45

ip
t

Table 1_new.docx

cr

Table 1. Simple descriptor for different molecules in commercial available lipid-based formulations together with the type of lipid

Alfacalcidol

Amprenavir

Bexarotene

Calcitrol

Ciprofloxacin

Dronabirol

Efavirenz

cLogP 1)

5.8

1.8

6.9

4.4

-0.8

5.9

4.5

Melting point (C) 2)

136

72-74

230-231

113

255-257

200

139-141

Hydrogen acceptors

Hydrogen donor

Polar surface area () 1)

40.5

131.2

37.3

60.9

72.9

29.5

38.3

Lipid formulation class

IV

III

III

used commercially 3)

ed

M
an

Compound

ce
pt

us

formulation class

1) Calculated using clogp from Daylight inc. (Laguna Niguel, CA , USA) and MOE, version 2008 (Chemical computing group, Montreal, Canada) for the PSA calculations

Ac

2) From www.drugbank.ca

3) Classified by Mullertz et al. (2010)

Page 39 of 45

ip
t
Fenofibrate

Ibudilast

Progesteron

cLogP 1)

5.3

3.7

4.1

Melting point (C) 2)

80.5

53-54

121

Hydrogen acceptors

Hydrogen donor

Polar surface area () 1)

52.6

34.4

Lipid formulation class

IV

Saquinavir Tiranavir

Valproic acid

Cyclosporine

3.2

7.8

2.8

3.4

78-82

350

86-89

120-130

151

23

34.1

145.8

166.8

105.5

37.3

278.8

IV

III + IV

III

ed

M
an

5.2

Ac

ce
pt

used commercially 3)

IV

Ritomair

us

Compound

cr

Table 1 cont.

Page 40 of 45

an

us

cr

graphical abstract.pdf

Drug

Ac

ce

pt

ed

In vitro lipolysis

Lipid-based formulations

Page 41 of 45

Figure 1.pdf

Figure 1

Drug

Ac
ce
p

Formation
of mixed
micelles

te

an

us

cr

ip
t

Lipid
digestion

Page 42 of 45

Figure 2.pdf

Figure 2

us

cr

ip
t

(A)

an

(B)
Lipid phase

Dispersion

80
60
40

100
80
60
40

20

20

II

140
120

Ac

% Fenofibrate

100

Digestion

ce
pt

120

Mass of dissolved Fenofibrate (mg)

(C)

ed

Pellet phase

Aqueous phase

III

II

III

Page 43 of 45

Figure 3.pdf

Figure 3

100

12:0/18:2/12:0

ip
t

10:0/18:2/10:0
2

us

8:0/18:2/8:0

cr

Cumulative % of MCFA in lymph

(A)

an

Time (h)

LCT

MCT
SCT
Lipid-free

Ac

ce
pt

ed

(B)

24

Page 44 of 45

Figure 4.pdf

Figure 4

us

cr

ip
t

(A)

Ac
ce
pt
e

an

(B)

Page 45 of 45

Das könnte Ihnen auch gefallen